1
|
George BM, Eleftheriou M, Yankova E, Perr J, Chai P, Nestola G, Almahayni K, Evans S, Damaskou A, Hemberger H, Lebedenko CG, Rak J, Yu Q, Bapcum E, Russell J, Bagri J, Volk RF, Spiekermann M, Stone RM, Giotopoulos G, Huntly BJP, Baxter J, Camargo F, Liu J, Zaro BW, Vassiliou GS, Möckl L, de la Rosa J, Flynn RA, Tzelepis K. Treatment of acute myeloid leukemia models by targeting a cell surface RNA-binding protein. Nat Biotechnol 2025:10.1038/s41587-025-02648-2. [PMID: 40269321 DOI: 10.1038/s41587-025-02648-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/20/2025] [Indexed: 04/25/2025]
Abstract
Immunotherapies for acute myeloid leukemia (AML) and other cancers are limited by a lack of tumor-specific targets. Here we discover that RNA-binding proteins and glycosylated RNAs (glycoRNAs) form precisely organized nanodomains on cancer cell surfaces. We characterize nucleophosmin (NPM1) as an abundant cell surface protein (csNPM1) on a variety of tumor types. With a focus on AML, we observe csNPM1 on blasts and leukemic stem cells but not on normal hematopoietic stem cells. We develop a monoclonal antibody to target csNPM1, which exhibits robust anti-tumor activity in multiple syngeneic and xenograft models of AML, including patient-derived xenografts, without observable toxicity. We find that csNPM1 is expressed in a mutation-agnostic manner on primary AML cells and may therefore offer a general strategy for detecting and treating AML. Surface profiling and in vivo work also demonstrate csNPM1 as a target on solid tumors. Our data suggest that csNPM1 and its neighboring glycoRNA-cell surface RNA-binding protein (csRBP) clusters may serve as an alternative antigen class for therapeutic targeting or cell identification.
Collapse
Affiliation(s)
- Benson M George
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria Eleftheriou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | - Eliza Yankova
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | - Jonathan Perr
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Peiyuan Chai
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Gianluca Nestola
- Department of Physics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Karim Almahayni
- Department of Physics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
- Max Planck Institute for the Science of Light, Erlangen, Germany
| | - Siân Evans
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Aristi Damaskou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Helena Hemberger
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Charlotta G Lebedenko
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Justyna Rak
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Qi Yu
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Ece Bapcum
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - James Russell
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Jaana Bagri
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Regan F Volk
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | | | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - George Giotopoulos
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Brian J P Huntly
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Joanna Baxter
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Fernando Camargo
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Jie Liu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Balyn W Zaro
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - George S Vassiliou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Leonhard Möckl
- Max Planck Institute for the Science of Light, Erlangen, Germany
- Faculty of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Faculty of Sciences, Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jorge de la Rosa
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Cambridge Institute of Science, Altos Labs, Cambridge, UK
| | - Ryan A Flynn
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| | - Konstantinos Tzelepis
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK.
- Wellcome Trust Sanger Institute, Hinxton, UK.
| |
Collapse
|
2
|
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
|
3
|
Liaki V, Rosas-Perez B, Guerra C. Unlocking the Genetic Secrets of Pancreatic Cancer: KRAS Allelic Imbalances in Tumor Evolution. Cancers (Basel) 2025; 17:1226. [PMID: 40227826 PMCID: PMC11987834 DOI: 10.3390/cancers17071226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) belongs to the types of cancer with the highest lethality. It is also remarkably chemoresistant to the few available cytotoxic therapeutic options. PDAC is characterized by limited mutational heterogeneity of the known driver genes, KRAS, CDKN2A, TP53, and SMAD4, observed in both early-stage and advanced tumors. In this review, we summarize the two proposed models of genetic evolution of pancreatic cancer. The gradual or stepwise accumulated mutations model has been widely studied. On the contrary, less evidence exists on the more recent simultaneous model, according to which rapid tumor evolution is driven by the concurrent accumulation of genetic alterations. In both models, oncogenic KRAS mutations are the main initiating event. Here, we analyze the emerging topic of KRAS allelic imbalances and how it arises during tumor evolution, as it is often detected in advanced and metastatic PDAC. We also summarize recent evidence on how it affects tumor biology, metastasis, and response to therapy. To this extent, we highlight the necessity to include studies of KRAS allelic frequencies in the design of future therapeutic strategies against pancreatic cancer.
Collapse
Affiliation(s)
- Vasiliki Liaki
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Blanca Rosas-Perez
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Carmen Guerra
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
4
|
Schulte L, Beck A, Marienfeld R, Azoitei N, Barth TFE, Beutel A, Benes V, Büchler MW, Gaisa NT, Kilani K, Giese N, Michalski CW, Möller P, Perkhofer L, Rausch T, Repky S, Roger E, Scheible J, Seufferlein T, Schirmacher P, Berger AW, Hackert T, Kleger A. Unveiling the intriguing relationship: oncogenic KRAS, morphological shifts, and mutational complexity in pancreatic mucinous cystic neoplasms. J Pathol 2025; 265:401-407. [PMID: 39906959 PMCID: PMC11880986 DOI: 10.1002/path.6397] [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: 05/16/2024] [Revised: 11/06/2024] [Accepted: 12/19/2024] [Indexed: 02/06/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) often arises from preexisting cystic lesions such as intraductal papillary mucinous neoplasms (IPMN) and mucinous cystic neoplasms (MCN). This study investigated the molecular heterogeneity and mutational landscape of MCN in relation to PDAC, highlighting the significance of KRAS mutations in tumor progression. Utilizing targeted next-generation sequencing on low-grade MCN and invasive PDAC samples, we identified a substantial overlap in mutational profiles, particularly mutations in KRAS, TP53, and FBXW7. Specifically, 69.2% of MCN exhibited somatic mutations, with KRAS mutations being a predominant oncogenic driver. The characterization of mutant versus wildtype KRAS variant allele frequencies (VAF) indicated higher mutation levels in PDAC compared to MCN, suggesting an evolutionary trajectory toward malignancy. Further histological analysis of 12 additional MCN cases revealed significant intratumor heterogeneity, with variant KRAS mutation distributions correlating with distinct cellular morphologies and dysplastic features. Additionally, we explored the potential of liquid biopsies, demonstrating a concordance rate of 71.4% for KRAS mutation detection in circulating tumor DNA (ctDNA) relative to tissue biopsies across cohorts. Our findings underscore the relevance of evaluating KRAS mutations-herein referred to as VAF per microdissected region-as they relate to histopathological markers of dysplasia, contributing to improved stratification of pancreatic lesions and facilitating personalized treatment strategies. In conclusion, this comprehensive analysis of MCN highlights the importance of KRAS as a crucial biomarker for both malignant progression and therapeutic decision-making in pancreatic pathology. Ultimately, our study suggests that characterizing the mutational landscape and histological features of MCN can enhance early detection and intervention strategies for at-risk patients. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lucas‐Alexander Schulte
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine 1Ulm University HospitalUlmGermany
| | - Annika Beck
- Department of PathologyUlm University HospitalUlmGermany
| | | | - Ninel Azoitei
- Institute of Molecular Oncology and Stem Cell Biology (IMOS)Ulm University HospitalUlmGermany
| | | | - Alica Beutel
- Department of Internal Medicine 1Ulm University HospitalUlmGermany
| | - Vladimir Benes
- Genomics Core FacilityEuropean Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Markus W Büchler
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | | | - Katja Kilani
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine 1Ulm University HospitalUlmGermany
| | - Nathalia Giese
- EPZ‐Biobank, Division of Pancreatic Cancer Research, Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Christoph W Michalski
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Peter Möller
- Department of PathologyUlm University HospitalUlmGermany
| | - Lukas Perkhofer
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine 1Ulm University HospitalUlmGermany
| | - Tobias Rausch
- Genomics Core FacilityEuropean Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Stefan Repky
- Institute of Epidemiology and Medical BiometryUlm UniversityUlmGermany
| | - Elodie Roger
- Institute of Molecular Oncology and Stem Cell Biology (IMOS)Ulm University HospitalUlmGermany
| | - Jeanette Scheible
- Institute of Molecular Oncology and Stem Cell Biology (IMOS)Ulm University HospitalUlmGermany
| | | | - Peter Schirmacher
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Andreas Wolfgang Berger
- Department of Internal Medicine 1Ulm University HospitalUlmGermany
- Evangelisches Krankenhaus Königin Elisabeth HerzbergeDepartment of GastroenterologyBerlinGermany
| | - Thilo Hackert
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
- Department of General SurgeryUniversity Hospital Hamburg EppendorfHamburgGermany
| | - Alexander Kleger
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine 1Ulm University HospitalUlmGermany
- Institute of Molecular Oncology and Stem Cell Biology (IMOS)Ulm University HospitalUlmGermany
| |
Collapse
|
5
|
Fiorini E, Malinova A, Schreyer D, Pasini D, Bevere M, Alessio G, Rosa D, D'Agosto S, Azzolin L, Milite S, Andreani S, Lupo F, Veghini L, Grimaldi S, Pedron S, Castellucci M, Nourse C, Salvia R, Malleo G, Ruzzenente A, Guglielmi A, Milella M, Lawlor RT, Luchini C, Agostini A, Carbone C, Pilarsky C, Sottoriva A, Scarpa A, Tuveson DA, Bailey P, Corbo V. MYC ecDNA promotes intratumour heterogeneity and plasticity in PDAC. Nature 2025; 640:811-820. [PMID: 40074906 PMCID: PMC12003172 DOI: 10.1038/s41586-025-08721-9] [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: 10/06/2023] [Accepted: 01/30/2025] [Indexed: 03/14/2025]
Abstract
Intratumour heterogeneity and phenotypic plasticity drive tumour progression and therapy resistance1,2. Oncogene dosage variation contributes to cell-state transitions and phenotypic heterogeneity3, thereby providing a substrate for somatic evolution. Nonetheless, the genetic mechanisms underlying phenotypic heterogeneity are still poorly understood. Here we show that extrachromosomal DNA (ecDNA) is a major source of high-level focal amplification in key oncogenes and a major contributor of MYC heterogeneity in pancreatic ductal adenocarcinoma (PDAC). We demonstrate that ecDNAs drive varying levels of MYC dosage, depending on their regulatory landscape, enabling cancer cells to rapidly and reversibly adapt to microenvironmental changes. In the absence of selective pressure, a high ecDNA copy number imposes a substantial fitness cost on PDAC cells. We also show that MYC dosage affects cell morphology and dependence of cancer cells on stromal niche factors. Our work provides a detailed analysis of ecDNAs in PDAC and describes a new genetic mechanism driving MYC heterogeneity in PDAC.
Collapse
Affiliation(s)
- Elena Fiorini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Antonia Malinova
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Daniel Schreyer
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Davide Pasini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Michele Bevere
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Giorgia Alessio
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Diego Rosa
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Sabrina D'Agosto
- ARC-Net Research Centre, University of Verona, Verona, Italy
- Human Technopole, Milan, Italy
| | - Luca Azzolin
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Salvatore Milite
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Silvia Andreani
- ARC-Net Research Centre, University of Verona, Verona, Italy
- Department of Biochemistry and Molecular Biology, University of Würzburg, Würzburg, Germany
| | - Francesca Lupo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Lisa Veghini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Sonia Grimaldi
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Serena Pedron
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Craig Nourse
- Cancer Research UK Beatson Institute, Glasgow, UK
- Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal
| | - Roberto Salvia
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona, Verona, Italy
| | - Giuseppe Malleo
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona, Verona, Italy
| | - Andrea Ruzzenente
- Department of Surgical Sciences, Division of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | - Alfredo Guglielmi
- Department of Surgical Sciences, Division of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | - Michele Milella
- Section of Medical Oncology, Department of Medicine, University of Verona, Verona, Italy
| | - Rita T Lawlor
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Antonio Agostini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Rome, Italy
| | - Carmine Carbone
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | - Andrea Sottoriva
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Aldo Scarpa
- ARC-Net Research Centre, University of Verona, Verona, Italy
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Peter Bailey
- School of Cancer Sciences, University of Glasgow, Glasgow, UK.
- Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal.
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy.
- ARC-Net Research Centre, University of Verona, Verona, Italy.
| |
Collapse
|
6
|
Perelli L, Zhang L, Mangiameli S, Giannese F, Mahadevan KK, Peng F, Citron F, Khan H, Le C, Gurreri E, Carbone F, Russell AJC, Soeung M, Lam TNA, Lundgren S, Marisetty S, Zhu C, Catania D, Mohamed AMT, Feng N, Augustine JJ, Sgambato A, Tortora G, Draetta GF, Tonon G, Futreal A, Giuliani V, Carugo A, Viale A, Kim MP, Heffernan TP, Wang L, Kalluri R, Cittaro D, Chen F, Genovese G. Evolutionary fingerprints of epithelial-to-mesenchymal transition. Nature 2025; 640:1083-1092. [PMID: 40044861 DOI: 10.1038/s41586-025-08671-2] [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: 07/11/2023] [Accepted: 01/17/2025] [Indexed: 04/13/2025]
Abstract
Mesenchymal plasticity has been extensively described in advanced epithelial cancers; however, its functional role in malignant progression is controversial1-5. The function of epithelial-to-mesenchymal transition (EMT) and cell plasticity in tumour heterogeneity and clonal evolution is poorly understood. Here we clarify the contribution of EMT to malignant progression in pancreatic cancer. We used somatic mosaic genome engineering technologies to trace and ablate malignant mesenchymal lineages along the EMT continuum. The experimental evidence clarifies the essential contribution of mesenchymal lineages to pancreatic cancer evolution. Spatial genomic analysis, single-cell transcriptomic and epigenomic profiling of EMT clarifies its contribution to the emergence of genomic instability, including events of chromothripsis. Genetic ablation of mesenchymal lineages robustly abolished these mutational processes and evolutionary patterns, as confirmed by cross-species analysis of pancreatic and other human solid tumours. Mechanistically, we identified that malignant cells with mesenchymal features display increased chromatin accessibility, particularly in the pericentromeric and centromeric regions, in turn resulting in delayed mitosis and catastrophic cell division. Thus, EMT favours the emergence of genomic-unstable, highly fit tumour cells, which strongly supports the concept of cell-state-restricted patterns of evolution, whereby cancer cell speciation is propagated to progeny within restricted functional compartments. Restraining the evolutionary routes through ablation of clones capable of mesenchymal plasticity, and extinction of the derived lineages, halts the malignant potential of one of the most aggressive forms of human cancer.
Collapse
Affiliation(s)
- Luigi Perelli
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Li Zhang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah Mangiameli
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | - Krishnan K Mahadevan
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fuduan Peng
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francesca Citron
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hania Khan
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney Le
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Enrico Gurreri
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Andrew J C Russell
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Melinda Soeung
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Truong Nguyen Anh Lam
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sebastian Lundgren
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sujay Marisetty
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cihui Zhu
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Desiree Catania
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alaa M T Mohamed
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ningping Feng
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jithesh Jose Augustine
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessandro Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Multiplex Spatial Imaging Facility, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giampaolo Tortora
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulio F Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giovanni Tonon
- Center for Omics Sciences, IRCCS San Raffaele Institute, Milan, Italy
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Virginia Giuliani
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael P Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy P Heffernan
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center, UT Health Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Davide Cittaro
- Center for Omics Sciences, IRCCS San Raffaele Institute, Milan, Italy.
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
7
|
Salas-Escabillas DJ, Hoffman MT, Brender SM, Moore JS, Wen HJ, Benitz S, Davis ET, Long D, Wombwell AM, Chianis ERD, Allen-Petersen BL, Steele NG, Sears RC, Matsumoto I, DelGiorno KE, Crawford HC. Tuft cells transdifferentiate to neural-like progenitor cells in the progression of pancreatic cancer. Dev Cell 2025; 60:837-852.e3. [PMID: 39721583 DOI: 10.1016/j.devcel.2024.12.003] [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: 04/19/2024] [Revised: 09/13/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is partly initiated through the transdifferentiation of acinar cells to metaplasia, which progresses to neoplasia and cancer. Tuft cells (TCs) are chemosensory cells not found in the normal pancreas but arise in cancer precursor lesions and diminish during progression to carcinoma. These metaplastic TCs (mTCs) suppress tumor progression through communication with the tumor microenvironment, but their fate during progression is unknown. To determine the fate of mTCs during PDA progression, we created a dual recombinase lineage trace model, wherein a pancreas-specific FlpO was used to induce tumorigenesis, while a tuft-cell specific Pou2f3CreERT/+ driver was used to induce expression of a tdTomato reporter. We found that mTCs in carcinoma transdifferentiate into neural-like progenitor cells (NRPs), a cell type associated with poor survival in patients. Using conditional knockout and overexpression systems, we found that Myc activity in mTCs is necessary and sufficient to induce this tuft-to-neuroendocrine transition (TNT).
Collapse
Affiliation(s)
- Daniel J Salas-Escabillas
- Cancer Biology, University of Michigan, Ann Arbor, MI, USA; Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Megan T Hoffman
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Sydney M Brender
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Jacee S Moore
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Hui-Ju Wen
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Simone Benitz
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Erick T Davis
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Daniel Long
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Allison M Wombwell
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Ella Rose D Chianis
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | | | - Nina G Steele
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | | | - Kathleen E DelGiorno
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Howard C Crawford
- Department of Surgery, Henry Ford Health, Detroit, MI, USA; Department of Pharmacology and Toxicology, Michigan State University, Lansing, MI, USA.
| |
Collapse
|
8
|
Yang H, Zhang L, Kang X, Si Y, Song P, Su X. Reaction Pathway Differentiation Enabled Fingerprinting Signal for Single Nucleotide Variant Detection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412680. [PMID: 39903775 PMCID: PMC11948007 DOI: 10.1002/advs.202412680] [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: 10/10/2024] [Revised: 01/17/2025] [Indexed: 02/06/2025]
Abstract
Accurate identification of single-nucleotide variants (SNVs) is paramount for disease diagnosis. Despite the facile design of DNA hybridization probes, their limited specificity poses challenges in clinical applications. Here, a differential reaction pathway probe (DRPP) based on a dynamic DNA reaction network is presented. DRPP leverages differences in reaction intermediate concentrations between SNV and WT groups, directing them into distinct reaction pathways. This generates a strong pulse-like signal for SNV and a weak unidirectional increase signal for wild-type (WT). Through the application of machine learning to fluorescence kinetic data analysis, the classification of SNV and WT signals is automated with an accuracy of 99.6%, significantly exceeding the 80.7% accuracy of conventional methods. Additionally, sensitivity for variant allele frequency (VAF) is enhanced down to 0.1%, representing a ten-fold improvement over conventional approaches. DRPP accurately identified D614G and N501Y SNVs in the S gene of SARS-CoV-2 variants in patient swab samples with accuracy over 99% (n = 82). It determined the VAF of ovarian cancer-related mutations KRAS-G12R, NRAS-G12C, and BRAF-V600E in both tissue and blood samples (n = 77), discriminating cancer patients and healthy individuals with significant difference (p < 0.001). The potential integration of DRPP into clinical diagnostics, along with rapid amplification techniques, holds promise for early disease diagnostics and personalized diagnostics.
Collapse
Affiliation(s)
- Huixiao Yang
- State Key Laboratory of Organic‐Inorganic CompositesBeijing Key Laboratory of BioprocessBeijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029China
| | - Linghao Zhang
- State Key Laboratory of Organic‐Inorganic CompositesBeijing Key Laboratory of BioprocessBeijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029China
| | - Xinmiao Kang
- State Key Laboratory of Organic‐Inorganic CompositesBeijing Key Laboratory of BioprocessBeijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029China
| | - Yunpei Si
- School of Biomedical EngineeringZhangjiang Institute for Advanced Study and National Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
| | - Ping Song
- School of Biomedical EngineeringZhangjiang Institute for Advanced Study and National Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
| | - Xin Su
- State Key Laboratory of Organic‐Inorganic CompositesBeijing Key Laboratory of BioprocessBeijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029China
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijing100191China
| |
Collapse
|
9
|
Qiao C, Wang L, Huang C, Jia Q, Bao W, Guo P, Tan D, Chen Z, Shi C, Rao Z, Zhang R, Wei W, Wang Z. Engineered Bacteria Manipulate Cysteine Metabolism to Boost Ferroptosis-Based Pancreatic Ductal Adenocarcinoma Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412982. [PMID: 39696900 DOI: 10.1002/adma.202412982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/18/2024] [Indexed: 12/20/2024]
Abstract
Cysteine metabolism is a key determinant of the defense against ferroptosis in pancreatic ductal adenocarcinoma (PDAC). Blocking cysteine metabolism may trigger potent ferroptosis in PDAC cells by generating lipid peroxides during tumor metabolic processes. However, current methods to limit cysteine availability fall short, failing to efficiently block cysteine metabolism due to inadequate tumor targeting and compensatory cysteine sources. Inspired by sulfur-metabolizing bacteria, synthetic biology to develop an engineered bacterium capable of directly depleting cysteine to block its metabolism is used. Acting as a living drug, these engineered bacteria colonize the tumor and continuously produce engineered cyst(e)inase enzyme (CGL) under the stimulation of tumor hypoxia. The CGL exhausts the substrate cysteine, completely impeding cysteine metabolism. This process dismantles the ferroptosis defense system in PDAC cells, triggers potent ferroptosis, and achieves efficient treatment. The results demonstrate that engineered bacteria designed for cysteine metabolism modulation possess unparalleled advantages in efficacy, persistence, and precision in blocking cysteine metabolism, making them highly suitable for effective ferroptosis treatment of PDAC.
Collapse
Affiliation(s)
- Chaoqiang Qiao
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
- Guangzhou Institute of Technology, Xidian University, Guangzhou, 510555, P. R. China
| | - Lexuan Wang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Chuting Huang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Qian Jia
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Weier Bao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Peilin Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Dengxu Tan
- Laboratory Animal Center, Air Force Medical University, Xi'an, 710032, P. R. China
| | - Zhuang Chen
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Changhong Shi
- Laboratory Animal Center, Air Force Medical University, Xi'an, 710032, P. R. China
| | - Zhiping Rao
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Ruili Zhang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhongliang Wang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| |
Collapse
|
10
|
Ems M, Brichkina A, Lauth M. A safe haven for cancer cells: tumor plus stroma control by DYRK1B. Oncogene 2025; 44:341-347. [PMID: 39863750 PMCID: PMC11790486 DOI: 10.1038/s41388-025-03275-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
The development of resistance remains one of the biggest challenges in clinical cancer patient care and it comprises all treatment modalities from chemotherapy to targeted or immune therapy. In solid malignancies, drug resistance is the result of adaptive processes occurring in cancer cells or the surrounding tumor microenvironment (TME). Future therapy attempts will therefore benefit from targeting both, tumor and stroma compartments and drug targets which affect both sides will be highly appreciated. In this review, we describe a seemingly paradoxical oncogenic mediator with this potential: The dual-specificity tyrosine-phosphorylation regulated kinase 1B (DYRK1B). DYRK1B promotes proliferative quiescence and yet is overexpressed or amplified in many hyperproliferative malignancies including ovarian cancer and pancreatic cancer. In particular the latter disease is a paradigmatic example for a therapy-recalcitrant and highly stroma-rich cancer entity. Here, recent evidence suggests that DYRK1B exerts its oncogenic features by installing a protective niche for cancer cells by directly affecting cancer cells but also the TME. Specifically, DYRK1B not only fosters cell-intrinsic processes like cell survival, chemoresistance, and disease recurrence, but it also upregulates TME and cancer cell-protective innate immune checkpoints and down-modulates anti-tumoral macrophage functionality. In this article, we outline the well-established cell-autonomous roles of DYRK1B and extend its importance to the TME and the control of the tumor immune stroma. In summary, DYRK1B appears as a single novel key player creating a safe haven for cancer cells by acting cell-intrinsically and-extrinsically, leading to the promotion of cancer cell survival, chemoresistance, and relapse. Thus, DYRK1B appears as an attractive drug target for future therapeutic approaches.
Collapse
Affiliation(s)
- Miriam Ems
- Department of Gastroenterology, Endocrinology and Metabolism, Center for Tumor and Immune Biology, Philipps University Marburg, Marburg, Germany
| | - Anna Brichkina
- Institute of Systems Immunology, Philipps University Marburg, Marburg, Germany
| | - Matthias Lauth
- Department of Gastroenterology, Endocrinology and Metabolism, Center for Tumor and Immune Biology, Philipps University Marburg, Marburg, Germany.
| |
Collapse
|
11
|
Varghese AM, Perry MA, Chou JF, Nandakumar S, Muldoon D, Erakky A, Zucker A, Fong C, Mehine M, Nguyen B, Basturk O, Balogun F, Kelsen DP, Brannon AR, Mandelker D, Vakiani E, Park W, Yu KH, Stadler ZK, Schattner MA, Jarnagin WR, Wei AC, Chakravarty D, Capanu M, Schultz N, Berger MF, Iacobuzio-Donahue CA, Bandlamudi C, O'Reilly EM. Clinicogenomic landscape of pancreatic adenocarcinoma identifies KRAS mutant dosage as prognostic of overall survival. Nat Med 2025; 31:466-477. [PMID: 39753968 PMCID: PMC11835752 DOI: 10.1038/s41591-024-03362-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/17/2024] [Indexed: 01/11/2025]
Abstract
Nearly all pancreatic adenocarcinomas (PDAC) are genomically characterized by KRAS exon 2 mutations. Most patients with PDAC present with advanced disease and are treated with cytotoxic therapy. Genomic biomarkers prognostic of disease outcomes have been challenging to identify. Herein leveraging a cohort of 2,336 patients spanning all disease stages, we characterize the genomic and clinical correlates of outcomes in PDAC. We show that a genomic subtype of KRAS wild-type tumors is associated with early disease onset, distinct somatic and germline features, and significantly better overall survival. Allelic imbalances at the KRAS locus are widespread. KRAS mutant allele dosage gains, observed in one in five (20%) KRAS-mutated diploid tumors, are correlated with advanced disease and demonstrate prognostic potential across disease stages. With the rapidly expanding landscape of KRAS targeting, our findings have potential implications for clinical practice and for understanding de novo and acquired resistance to RAS therapeutics.
Collapse
Affiliation(s)
- Anna M Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Maria A Perry
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Joanne F Chou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Subhiksha Nandakumar
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Daniel Muldoon
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Amanda Erakky
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Amanda Zucker
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Christopher Fong
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Miika Mehine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Bastien Nguyen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Olca Basturk
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Fiyinfolu Balogun
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - David P Kelsen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - A Rose Brannon
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Diana Mandelker
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Efsevia Vakiani
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Kenneth H Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Mark A Schattner
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - William R Jarnagin
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Alice C Wei
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Debyani Chakravarty
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Nikolaus Schultz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Michael F Berger
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Christine A Iacobuzio-Donahue
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Chaitanya Bandlamudi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| |
Collapse
|
12
|
Tapia Contreras C, Falke JD, Seifert D, Schneider C, Krauß L, Fang X, Müller D, Demirdizen E, Spitzner M, De Oliveira T, Schneeweis C, Gaedcke J, Kaulfuß S, Mirzakhani K, Wollnik B, Conrads K, Beißbarth T, Salinas G, Hügel J, Beyer N, Rheinländer S, Sax U, Wirth M, Conradi L, Reichert M, Ellenrieder V, Ströbel P, Ghadimi M, Grade M, Saur D, Hessmann E, Schneider G. KRAS G 12C-inhibitor-based combination therapies for pancreatic cancer: insights from drug screening. Mol Oncol 2025; 19:295-310. [PMID: 39253995 PMCID: PMC11792994 DOI: 10.1002/1878-0261.13725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 06/06/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has limited treatment options, emphasizing the urgent need for effective therapies. The predominant driver in PDAC is mutated KRAS proto-oncogene, KRA, present in 90% of patients. The emergence of direct KRAS inhibitors presents a promising avenue for treatment, particularly those targeting the KRASG12C mutated allele, which show encouraging results in clinical trials. However, the development of resistance necessitates exploring potent combination therapies. Our objective was to identify effective KRASG12C-inhibitor combination therapies through unbiased drug screening. Results revealed synergistic effects with son of sevenless homolog 1 (SOS1) inhibitors, tyrosine-protein phosphatase non-receptor type 11 (PTPN11)/Src homology region 2 domain-containing phosphatase-2 (SHP2) inhibitors, and broad-spectrum multi-kinase inhibitors. Validation in a novel and unique KRASG12C-mutated patient-derived organoid model confirmed the described hits from the screening experiment. Our findings propose strategies to enhance KRASG12C-inhibitor efficacy, guiding clinical trial design and molecular tumor boards.
Collapse
Affiliation(s)
| | - Jonas Dominik Falke
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Dana‐Magdalena Seifert
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Carolin Schneider
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Lukas Krauß
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Xin Fang
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Denise Müller
- Institute of PathologyUniversity Medical CenterGöttingenGermany
| | - Engin Demirdizen
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Melanie Spitzner
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Tiago De Oliveira
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Christian Schneeweis
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University MunichGermany
| | - Jochen Gaedcke
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
| | - Silke Kaulfuß
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Institute of Human GeneticsUniversity Medical Center GöttingenGermany
| | - Kimia Mirzakhani
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Institute of Human GeneticsUniversity Medical Center GöttingenGermany
| | - Bernd Wollnik
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Institute of Human GeneticsUniversity Medical Center GöttingenGermany
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (MBExC)University of GöttingenGermany
| | - Karly Conrads
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Department of Medical BioinformaticsUniversity Medical Center GöttingenGermany
| | - Tim Beißbarth
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Department of Medical BioinformaticsUniversity Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
- Campus‐Institute Data Science (CIDAS)GöttingenGermany
| | - Gabriela Salinas
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- NGS Integrative Genomics Core Unit (NIG)University Medical Center Göttingen (UMG)Germany
| | - Jonas Hügel
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Department of Medical InformaticsUniversity Medical CenterGöttingenGermany
| | - Nils Beyer
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Department of Medical InformaticsUniversity Medical CenterGöttingenGermany
| | - Sophia Rheinländer
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Department of Medical InformaticsUniversity Medical CenterGöttingenGermany
| | - Ulrich Sax
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Campus‐Institute Data Science (CIDAS)GöttingenGermany
- Department of Medical InformaticsUniversity Medical CenterGöttingenGermany
| | - Matthias Wirth
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- Department of Hematology, Oncology and Cancer ImmunologyCampus Benjamin Franklin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinGermany
| | - Lena‐Christin Conradi
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
| | - Maximilian Reichert
- Medical Clinic and Polyclinic II, Klinikum rechts der IsarTechnical University MunichGermany
- Translational Pancreatic Research Cancer Center, Medical Clinic and Polyclinic II, Klinikum rechts der IsarTechnical University MunichGermany
- Center for Protein Assemblies (CPA)Technical University of MunichGarchingGermany
- Center for Organoid Systems and Tissue Engineering (COS)Technical University MunichGarchingGermany
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership Between DKFZ and University Hospital Klinikum rechts der IsarMunichGermany
| | - Volker Ellenrieder
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
- Department of Gastroenterology, Gastrointestinal Oncology and EndocrinologyUniversity Medical Center GöttingenGermany
| | - Philipp Ströbel
- Institute of PathologyUniversity Medical CenterGöttingenGermany
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
| | - Marian Grade
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
| | - Dieter Saur
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University MunichGermany
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership Between DKFZ and University Hospital Klinikum rechts der IsarMunichGermany
| | - Elisabeth Hessmann
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
- Department of Gastroenterology, Gastrointestinal Oncology and EndocrinologyUniversity Medical Center GöttingenGermany
| | - Günter Schneider
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University MunichGermany
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
| |
Collapse
|
13
|
Yun WG, Kim D, Han Y, Kwon W, Lee SG, Jang JY, Park D. Multiomic quantification of the KRAS mutation dosage improves the preoperative prediction of survival and recurrence in patients with pancreatic ductal adenocarcinoma. Exp Mol Med 2025; 57:193-203. [PMID: 39779977 PMCID: PMC11799340 DOI: 10.1038/s12276-024-01382-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/29/2024] [Accepted: 10/25/2024] [Indexed: 01/11/2025] Open
Abstract
Most cancer mutation profiling studies are laboratory-based and lack direct clinical application. For clinical use, it is necessary to focus on key genes and integrate them with relevant clinical variables. We aimed to evaluate the prognostic value of the dosage of the KRAS G12 mutation, a key pancreatic ductal adenocarcinoma (PDAC) variant and to investigate the biological mechanism of the prognosis associated with the dosage of the KRAS G12 mutation. In this retrospective cohort study, we analyzed 193 surgically treated patients with PDAC between 2009 and 2016. RNA, whole-exome, and KRAS-targeted sequencing data were used to estimate the dosage of the KRAS G12 mutant. Our prognostic scoring system included the mutation dosage from targeted sequencing ( > 0.195, 1 point), maximal tumor diameter at preoperative imaging ( > 20 mm, 1 point), and carbohydrate antigen 19-9 levels ( > 150 U/mL, 1 point). The KRAS mutation dosage exhibited comparable performance with clinical variables for survival prediction. High KRAS mutation dosages activated the cell cycle, leading to high mutation rates and poor prognosis. According to prognostic scoring systems that integrate mutation dosage with clinical factors, patients with 0 points had superior median overall survival of 97.0 months and 1-year, 3-year, and 5-year overall survival rates of 95.8%, 70.8%, and 66.4%, respectively. In contrast, patients with 3 points had worse median overall survival of only 16.0 months and 1-year, 3-year, and 5-year overall survival rates of 65.2%, 8.7%, and 8.7%, respectively. The incorporation of the KRAS G12 mutation dosage variable into prognostic scoring systems can improve clinical variable-based survival prediction, highlighting the feasibility of an integrated scoring system with clinical significance.
Collapse
Affiliation(s)
- Won-Gun Yun
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Daeun Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- Ajou Energy Science Research Center, Ajou University, Suwon, South Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Seong-Geun Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| | - Daechan Park
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea.
- Advanced College of Bio-convergence Engineering, Ajou University, Suwon, South Korea.
| |
Collapse
|
14
|
Fey SK, Najumudeen AK, Watt DM, Millett LM, Ford CA, Gilroy K, Simpson RJ, McLay K, Upstill-Goddard R, Chang D, Clark W, Nixon C, Birch JL, Barry ST, Morton JP, Campbell AD, Sansom OJ. KRAS Loss of Heterozygosity Promotes MAPK-Dependent Pancreatic Ductal Adenocarcinoma Initiation and Induces Therapeutic Sensitivity to MEK Inhibition. Cancer Res 2025; 85:251-262. [PMID: 39412982 PMCID: PMC11733531 DOI: 10.1158/0008-5472.can-23-2709] [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: 09/06/2023] [Revised: 07/11/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
Pancreatic cancer is characterized by the prevalence of oncogenic mutations in KRAS. Previous studies have reported that altered KRAS gene dosage drives progression and metastasis in pancreatic cancer. Whereas the role of oncogenic KRAS mutations is well characterized, the relevance of the partnering wild-type (WT) KRAS allele in pancreatic cancer is less well understood and controversial. Using in vivo mouse modeling of pancreatic cancer, we demonstrated that WT KRAS restrains the oncogenic impact of mutant KRAS and dramatically impacts both KRAS-mediated tumorigenesis and therapeutic response. Mechanistically, deletion of WT Kras increased oncogenic KRAS signaling through the downstream MAPK effector pathway, driving pancreatic intraepithelial neoplasia initiation. In addition, in the KPC mouse model, a more aggressive model of pancreatic cancer, lack of WT KRAS led to accelerated initiation but delayed tumor progression. These tumors had altered stroma and an enrichment of immunogenic gene signatures. Importantly, loss of WT Kras sensitized Kras mutant tumors to MEK1/2 inhibition though tumors eventually became resistant and then rapidly progressed. This study demonstrates the repressive role of WT KRAS during pancreatic tumorigenesis and highlights the critical impact of the presence of WT KRAS in both tumor progression and therapeutic response in pancreatic cancer. Significance: KRAS allelic status impacts pancreatic cancer progression and has the potential to guide effective treatment in a substantial subset of patients.
Collapse
Affiliation(s)
- Sigrid K. Fey
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Dale M. Watt
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
| | - Laura M. Millett
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Kathryn Gilroy
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
| | | | - Kathy McLay
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Rosanna Upstill-Goddard
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Wolfson Wohl Cancer Research Centre, Glasgow, United Kingdom
| | - David Chang
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Wolfson Wohl Cancer Research Centre, Glasgow, United Kingdom
| | - William Clark
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
| | - Colin Nixon
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
| | - Joanna L. Birch
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Simon T. Barry
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Jennifer P. Morton
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Owen J. Sansom
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
15
|
Ying H, Kimmelman AC, Bardeesy N, Kalluri R, Maitra A, DePinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 2025; 39:36-63. [PMID: 39510840 PMCID: PMC11789498 DOI: 10.1101/gad.351863.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a grim prognosis for patients. Recent multidisciplinary research efforts have provided critical insights into its genetics and tumor biology, creating the foundation for rational development of targeted and immune therapies. Here, we review the PDAC genomic landscape and the role of specific oncogenic events in tumor initiation and progression, as well as their contributions to shaping its tumor biology. We further summarize and synthesize breakthroughs in single-cell and metabolic profiling technologies that have illuminated the complex cellular composition and heterotypic interactions of the PDAC tumor microenvironment, with an emphasis on metabolic cross-talk across cancer and stromal cells that sustains anabolic growth and suppresses tumor immunity. These conceptual advances have generated novel immunotherapy regimens, particularly cancer vaccines, which are now in clinical testing. We also highlight the advent of KRAS targeted therapy, a milestone advance that has transformed treatment paradigms and offers a platform for combined immunotherapy and targeted strategies. This review provides a perspective summarizing current scientific and therapeutic challenges as well as practice-changing opportunities for the PDAC field at this major inflection point.
Collapse
Affiliation(s)
- Haoqiang Ying
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA;
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Alec C Kimmelman
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, New York 10016, USA
- Department of Radiation Oncology, New York University Grossman School of Medicine, New York, New York 10016, USA
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts 02114, USA
- The Cancer Program, Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Raghu Kalluri
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Anirban Maitra
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Sheikh Ahmed Pancreatic Cancer Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ronald A DePinho
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA;
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
16
|
Pian LL, Song MH, Wang TF, Qi L, Peng TL, Xie KP. Identification and analysis of pancreatic intraepithelial neoplasia: opportunities and challenges. Front Endocrinol (Lausanne) 2025; 15:1401829. [PMID: 39839479 PMCID: PMC11746065 DOI: 10.3389/fendo.2024.1401829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Pancreatic intraepithelial neoplasia (PanIN) is the most common precursor lesion of pancreatic ductal adenocarcinoma (PDAC), which has poor prognosis with a short median overall survival of 6-12 months and a low 5-year survival rate of approximately 3%. It is crucial to remove PanIN lesions to prevent the development of invasive PDAC, as PDAC spreads rapidly outside the pancreas. This review aims to provide the latest knowledge on PanIN risk, pathology, cellular origin, genetic susceptibility, and diagnosis, while identifying research gaps that require further investigation in this understudied area of precancerous lesions. PanINs are classified into PanIN 1, PanIN 2, and PanIN 3, with PanIN 3 having the highest likelihood of developing into invasive PDAC. Differentiating between PanIN 2 and PanIN 3 is clinically significant. Genetic alterations found in PDAC are also present in PanIN and increase with the grade of PanIN. Imaging methods alone are insufficient for distinguishing PanIN, necessitating the use of genetic and molecular tests for identification. In addition, metabolomics technologies and miRNAs are playing an increasingly important role in the field of cancer diagnosis, offering more possibilities for efficient identification of PanIN. Although detecting and stratifying the risk of PanIN poses challenges, the combined utilization of imaging, genetics, and metabolomics holds promise for improving patient survival in this field.
Collapse
Affiliation(s)
- Ling-ling Pian
- School of Medicine, The South China University of Technology, Guangzhou, Guangdong, China
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Mei-hui Song
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Teng-fei Wang
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Ling Qi
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Tie-li Peng
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Ke-ping Xie
- School of Medicine, The South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Papargyriou A, Najajreh M, Cook DP, Maurer CH, Bärthel S, Messal HA, Ravichandran SK, Richter T, Knolle M, Metzler T, Shastri AR, Öllinger R, Jasper J, Schmidleitner L, Wang S, Schneeweis C, Ishikawa-Ankerhold H, Engleitner T, Mataite L, Semina M, Trabulssi H, Lange S, Ravichandra A, Schuster M, Mueller S, Peschke K, Schäfer A, Dobiasch S, Combs SE, Schmid RM, Bausch AR, Braren R, Heid I, Scheel CH, Schneider G, Zeigerer A, Luecken MD, Steiger K, Kaissis G, van Rheenen J, Theis FJ, Saur D, Rad R, Reichert M. Heterogeneity-driven phenotypic plasticity and treatment response in branched-organoid models of pancreatic ductal adenocarcinoma. Nat Biomed Eng 2024:10.1038/s41551-024-01273-9. [PMID: 39658630 DOI: 10.1038/s41551-024-01273-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 09/26/2024] [Indexed: 12/12/2024]
Abstract
In patients with pancreatic ductal adenocarcinoma (PDAC), intratumoural and intertumoural heterogeneity increases chemoresistance and mortality rates. However, such morphological and phenotypic diversities are not typically captured by organoid models of PDAC. Here we show that branched organoids embedded in collagen gels can recapitulate the phenotypic landscape seen in murine and human PDAC, that the pronounced molecular and morphological intratumoural and intertumoural heterogeneity of organoids is governed by defined transcriptional programmes (notably, epithelial-to-mesenchymal plasticity), and that different organoid phenotypes represent distinct tumour-cell states with unique biological features in vivo. We also show that phenotype-specific therapeutic vulnerabilities and modes of treatment-induced phenotype reprogramming can be captured in phenotypic heterogeneity maps. Our methodology and analyses of tumour-cell heterogeneity in PDAC may guide the development of phenotype-targeted treatment strategies.
Collapse
Affiliation(s)
- Aristeidis Papargyriou
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Mulham Najajreh
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
| | - David P Cook
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Carlo H Maurer
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Stefanie Bärthel
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Chair for Translational Cancer Research and Institute of Experimental Cancer Therapy, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Hendrik A Messal
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sakthi K Ravichandran
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
| | - Till Richter
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
- Department of Mathematics, School of Computing, Information and Technology, Technical University of Munich, Munich, Germany
| | - Moritz Knolle
- Institute of Diagnostic and Interventional Radiology, Klinikum rechts der Isar München, Technical University of Munich, Munich, Germany
- Artificial Intelligence in Medicine and Healthcare, Technical University of Munich, Munich, Germany
| | - Thomas Metzler
- Comparative Experimental Pathology, Institut für Allgemeine Pathologie und Pathologische Anatomie, School of Medicine, Technical University of Munich, Munich, Germany
| | - Akul R Shastri
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Rupert Öllinger
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jacob Jasper
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
| | - Laura Schmidleitner
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Surui Wang
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Christian Schneeweis
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Chair for Translational Cancer Research and Institute of Experimental Cancer Therapy, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Hellen Ishikawa-Ankerhold
- Department of Medicine I, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Thomas Engleitner
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Laura Mataite
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
| | - Mariia Semina
- Institute of Diagnostic and Interventional Radiology, Klinikum rechts der Isar München, Technical University of Munich, Munich, Germany
| | - Hussein Trabulssi
- Institute of Diagnostic and Interventional Radiology, Klinikum rechts der Isar München, Technical University of Munich, Munich, Germany
| | - Sebastian Lange
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Aashreya Ravichandra
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Maximilian Schuster
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
| | - Sebastian Mueller
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Katja Peschke
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
| | - Arlett Schäfer
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
| | - Sophie Dobiasch
- Department of Radiation Oncology, Technical University of Munich, Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Technical University of Munich, Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Roland M Schmid
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Andreas R Bausch
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
- Lehrstuhl für Zell Biophysik E27, Physik Department, Technische Universität München, Garching, Germany
| | - Rickmer Braren
- Institute of Diagnostic and Interventional Radiology, Klinikum rechts der Isar München, Technical University of Munich, Munich, Germany
| | - Irina Heid
- Institute of Diagnostic and Interventional Radiology, Klinikum rechts der Isar München, Technical University of Munich, Munich, Germany
| | - Christina H Scheel
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
- Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - Günter Schneider
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Malte D Luecken
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), München, Germany
| | - Katja Steiger
- Comparative Experimental Pathology, Institut für Allgemeine Pathologie und Pathologische Anatomie, School of Medicine, Technical University of Munich, Munich, Germany
| | - Georgios Kaissis
- Institute of Diagnostic and Interventional Radiology, Klinikum rechts der Isar München, Technical University of Munich, Munich, Germany
- Artificial Intelligence in Medicine and Healthcare, Technical University of Munich, Munich, Germany
- Institute for Machine Learning in Biomedical Imaging, Helmholtz Zentrum München, München, Germany
- Department of Computing, Imperial College London, London, UK
- Munich Center for Machine Learning (MCML), München, Germany
- School of Computation, Information and Technology, Technische Universität München, München, Germany
| | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
- Department of Mathematics, School of Computing, Information and Technology, Technical University of Munich, Munich, Germany
- Cellular Genetics Programme, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Dieter Saur
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Chair for Translational Cancer Research and Institute of Experimental Cancer Therapy, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Roland Rad
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Maximilian Reichert
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany.
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, München, Germany.
- Center for Functional Protein Assemblies, Technical University of Munich, Garching, Germany.
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany.
- Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich, Munich, Germany.
| |
Collapse
|
18
|
Veghini L, Pasini D, Fang R, Delfino P, Filippini D, Neander C, Vicentini C, Fiorini E, Lupo F, D'Agosto SL, Carbone C, Agostini A, Piro G, Rosa D, Bevere M, Markus P, Behrens D, Luchini C, Lawlor RT, Scarpa A, Biffi G, Cheung PF, Siveke JT, Corbo V. Differential activity of MAPK signalling defines fibroblast subtypes in pancreatic cancer. Nat Commun 2024; 15:10534. [PMID: 39627211 PMCID: PMC11615044 DOI: 10.1038/s41467-024-54975-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
Fibroblast heterogeneity is increasingly recognised across cancer conditions. Given their important contribution to disease progression, mapping fibroblasts' heterogeneity is critical to devise effective anti-cancer therapies. Cancer-associated fibroblasts (CAFs) represent the most abundant cell population in pancreatic ductal adenocarcinoma (PDAC). Whether CAF phenotypes are differently specified by PDAC cell lineages remains to be elucidated. Here, we reveal an important role for the MAPK signalling pathway in defining PDAC CAF phenotypes. We show that epithelial MAPK activity promotes the myofibroblastic differentiation of CAFs by sustaining the expression and secretion of TGF-β1. We integrate single-cell profiling of post-perturbation transcriptional responses from mouse models with cellular and spatial profiles of human tissues to define a MAPKhigh CAF (mapCAF) phenotype. We show that this phenotype associates with basal-like tumour cells and reduced frequency of CD8+ T cells. In addition to elevated MAPK activity, this mapCAF phenotype is characterized by TGF-β signalling, hypoxia responsive signatures, and immunoregulatory gene programs. Furthermore, the mapCAF signature is enriched in myofibroblastic CAFs from various cancer conditions and correlates with reduced response to immune checkpoint inhibition in melanoma. Altogether, our data expand our knowledge on CAF phenotype heterogeneity and reveal a potential strategy for targeting myofibroblastic CAFs in vivo.
Collapse
Affiliation(s)
- Lisa Veghini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Davide Pasini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Rui Fang
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK), Partner Site Essen, A Partnership Between German Cancer Research Center (DKFZ) and University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Pietro Delfino
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- Department of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Dea Filippini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Christian Neander
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK), Partner Site Essen, A Partnership Between German Cancer Research Center (DKFZ) and University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Caterina Vicentini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Elena Fiorini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Francesca Lupo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Sabrina L D'Agosto
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- Human Technopole, Milan, Italy
| | - Carmine Carbone
- Department of Medical and Surgical Sciences, Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Agostini
- Department of Medical and Surgical Sciences, Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Geny Piro
- Department of Medical and Surgical Sciences, Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Diego Rosa
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Michele Bevere
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Peter Markus
- Department of General, Visceral, and Trauma Surgery, Elisabeth Hospital Essen, Essen, Germany
| | - Diana Behrens
- EPO-Experimental Pharmacology and Oncology GmbH, Berlin, Germany
| | - Claudio Luchini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Rita T Lawlor
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Giulia Biffi
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Phyllis F Cheung
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK), Partner Site Essen, A Partnership Between German Cancer Research Center (DKFZ) and University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Spatiotemporal Tumor Heterogeneity, DKTK, Partner Site Essen, A Partnership Between DKFZ and University Hospital Essen, Essen, Germany
| | - Jens T Siveke
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK), Partner Site Essen, A Partnership Between German Cancer Research Center (DKFZ) and University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy.
| |
Collapse
|
19
|
Wang J, Du J, Luo X, Guo L, Liu Y, Zhou J, Zou Y, Lu Z, Pan X, Chen X, Zhong A, Wan X, Wang L, Liu H, Dai S, Zhang S, Xiong X, Tan P, Wang M, Wu B, Zhang Q, Wang Y, Zhang M, Lu R, Lin H, Li Y, Li Y, Han Z, Chen L, Hu B, Liu Y, Na F, Chen C. A platform of functional studies of ESCC-associated gene mutations identifies the roles of TGFBR2 in ESCC progression and metastasis. Cell Rep 2024; 43:114952. [PMID: 39527477 DOI: 10.1016/j.celrep.2024.114952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/31/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Genomics studies have detected numerous genetic alterations in esophageal squamous cell carcinoma (ESCC). However, the functions of these mutations largely remain elusive, partially due to a lack of feasible animal models. Here, we report a convenient platform with CRISPR-Cas9-mediated introduction of genetic alterations and orthotopic transplantation to generate a series of primary ESCC models in mice. With this platform, we validate multiple frequently mutated genes, including EP300, FAT1/2/4, KMT2D, NOTCH2, and TGFBR2, as tumor-suppressor genes in ESCC. Among them, TGFBR2 loss dramatically promotes tumorigenesis and multi-organ metastasis. Paradoxically, TGFBR2 deficiency leads to Smad3 activation, and disruption of Smad3 partially restrains the progression of Tgfbr2-mutated tumors. Drug screening with tumor organoids identifies that pinaverium bromide represses Smad3 activity and restrains Tgfbr2-deficient ESCC. Our studies provide a highly efficient platform to investigate the in vivo functions of ESCC-associated mutations and develop potential treatments for this miserable malignancy.
Collapse
Affiliation(s)
- Jian Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiajia Du
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiangmeng Luo
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linjie Guo
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yixin Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianfeng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yang Zou
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenghao Lu
- Chengdu OrganoidMed Medical Laboratory, West China Health Valley, Chengdu, Sichuan 610041, China
| | - Xiangyu Pan
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuelan Chen
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ailing Zhong
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xudong Wan
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongyu Liu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Siqi Dai
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shiyu Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingyu Xiong
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Tan
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Manli Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Baohong Wu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qi Zhang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yingjie Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mengsha Zhang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Runda Lu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huahang Lin
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Li
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yaxin Li
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zongkai Han
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Bing Hu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yu Liu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Feifei Na
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Chong Chen
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, No387-201 Hemin st., Chengdu, Sichuan 610212, China; Children's Medicine Key Laboratory of Sichuan Province, Sichuan 610041, China.
| |
Collapse
|
20
|
Benitz S, Steep A, Nasser MM, Preall J, Mahajan UM, McQuithey H, Loveless I, Davis ET, Wen HJ, Long DW, Metzler T, Zwernik S, Louw M, Rempinski D, Salas-Escabillas DJ, Brender SM, Song L, Huang L, Theisen BK, Zhang Z, Steele NG, Regel I, Bednar F, Crawford HC. ROR2 Regulates Cellular Plasticity in Pancreatic Neoplasia and Adenocarcinoma. Cancer Discov 2024; 14:2162-2182. [PMID: 38975886 PMCID: PMC11528200 DOI: 10.1158/2159-8290.cd-24-0137] [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: 01/25/2024] [Revised: 05/06/2024] [Accepted: 06/20/2024] [Indexed: 07/09/2024]
Abstract
Cellular plasticity is a hallmark of pancreatic ductal adenocarcinoma (PDAC) starting from the conversion of normal cells into precancerous lesions, to the progression of carcinoma subtypes associated with aggressiveness and therapeutic response. We discovered that normal acinar cell differentiation, maintained by the transcription factor PDX1, suppresses a broad gastric cell identity that is maintained in metaplasia, neoplasia, and the classical subtype of PDAC in a mouse and human. We identified the receptor tyrosine kinase ROR2 as marker of a gastric metaplasia-like identity in pancreas neoplasms. Ablation of Ror2 in a mouse model of pancreatic tumorigenesis promoted a switch to a gastric pit cell identity that largely persisted through progression to the classical subtype of PDAC. In both human and mouse pancreatic cancer, ROR2 activity continued to antagonize the gastric pit cell identity, strongly promoting an epithelial to mesenchymal transition, conferring resistance to KRAS inhibition, and vulnerability to AKT inhibition. Significance: We discovered the receptor tyrosine kinase ROR2 as an important regulator of cellular identity in pancreatic precancerous lesions and pancreatic cancer. ROR2 drives an aggressive PDAC phenotype and confers resistance to KRAS inhibitors, suggesting that targeting ROR2 will enhance sensitivity to this new generation of targeted therapies. See related commentary by Marasco and Misale, p. 2018.
Collapse
Affiliation(s)
- Simone Benitz
- Department of Surgery, Henry Ford Health, Detroit, Michigan
| | - Alec Steep
- Center of Translational Data Science, University of Chicago, Chicago, Illinois
| | | | - Jonathan Preall
- Cold Spring Harbor Laboratory Cancer Center, Cold Spring Harbor, New York
| | | | | | - Ian Loveless
- Department of Public Health Sciences, Henry Ford Health, Detroit, Michigan
| | - Erick T. Davis
- Department of Surgery, Henry Ford Health, Detroit, Michigan
| | - Hui-Ju Wen
- Department of Surgery, Henry Ford Health, Detroit, Michigan
| | - Daniel W. Long
- Department of Surgery, Henry Ford Health, Detroit, Michigan
| | - Thomas Metzler
- Comparative Experimental Pathology (CEP), Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Samuel Zwernik
- Department of Surgery, Henry Ford Health, Detroit, Michigan
| | - Michaela Louw
- Department of Surgery, Henry Ford Health, Detroit, Michigan
| | | | | | | | - Linghao Song
- Center of Translational Data Science, University of Chicago, Chicago, Illinois
| | - Ling Huang
- Department of Surgery, Henry Ford Health, Detroit, Michigan
| | | | - Zhenyu Zhang
- Center of Translational Data Science, University of Chicago, Chicago, Illinois
| | - Nina G. Steele
- Department of Surgery, Henry Ford Health, Detroit, Michigan
- Department of Pathology, Wayne State University, Detroit, Michigan
- Department of Pharmacology and Toxicology, Michigan State University, Lansing, Michigan
- Department of Oncology, Wayne State University, Detroit, Michigan
| | - Ivonne Regel
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Filip Bednar
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Howard C. Crawford
- Department of Surgery, Henry Ford Health, Detroit, Michigan
- Department of Pharmacology and Toxicology, Michigan State University, Lansing, Michigan
- Department of Oncology, Wayne State University, Detroit, Michigan
| |
Collapse
|
21
|
Singhal A, Styers HC, Rub J, Li Z, Torborg SR, Kim JY, Grbovic-Huezo O, Feng H, Tarcan ZC, Ozkan HS, Hallin J, Basturk O, Yaeger R, Christensen JG, Betel D, Yan Y, Chio IIC, de Stanchina E, Tammela T. A Classical Epithelial State Drives Acute Resistance to KRAS Inhibition in Pancreatic Cancer. Cancer Discov 2024; 14:2122-2134. [PMID: 38975873 PMCID: PMC11624508 DOI: 10.1158/2159-8290.cd-24-0740] [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: 05/22/2024] [Revised: 06/25/2024] [Accepted: 07/05/2024] [Indexed: 07/09/2024]
Abstract
Intratumoral heterogeneity in pancreatic ductal adenocarcinoma (PDAC) is characterized by a balance between basal and classical epithelial cancer cell states, with basal dominance associating with chemoresistance and a dismal prognosis. Targeting oncogenic KRAS, the primary driver of pancreatic cancer, shows early promise in clinical trials, but efficacy is limited by acquired resistance. Using genetically engineered mouse models and patient-derived xenografts, we find that basal PDAC cells are highly sensitive to KRAS inhibitors. Employing fluorescent and bioluminescent reporter systems, we longitudinally track cell-state dynamics in vivo and reveal a rapid, KRAS inhibitor-induced enrichment of the classical state. Lineage tracing uncovers that these enriched classical PDAC cells are a reservoir for disease relapse. Genetic or chemotherapy-mediated ablation of the classical cell state is synergistic with KRAS inhibition, providing a preclinical proof of concept for this therapeutic strategy. Our findings motivate combining classical state-directed therapies with KRAS inhibitors to deepen responses and counteract resistance in pancreatic cancer. Significance: KRAS inhibitors hold promise in pancreatic cancer, but responses are limited by acquired resistance. We find that a classical epithelial cancer cell state is acutely resistant to KRAS inhibition and serves as a reservoir for disease relapse. Targeting the classical state alongside KRAS inhibition deepens responses, revealing a potent therapeutic strategy. See related commentary by Marasco and Misale, p. 2018.
Collapse
Affiliation(s)
- Anupriya Singhal
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- David M. Rubenstein Center for Pancreatic Cancer, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hannah C. Styers
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jonathan Rub
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Zhuxuan Li
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, New York, New York, USA
| | - Stefan R. Torborg
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Jung Yun Kim
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Olivera Grbovic-Huezo
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Huijin Feng
- Institute for Cancer Genetics, Department of Genetics and Development and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Zeynep Cagla Tarcan
- David M. Rubenstein Center for Pancreatic Cancer, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hulya Sahin Ozkan
- David M. Rubenstein Center for Pancreatic Cancer, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jill Hallin
- Mirati Therapeutics, San Diego, California 92121, USA
| | - Olca Basturk
- David M. Rubenstein Center for Pancreatic Cancer, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rona Yaeger
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Doron Betel
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Division of Hematology & Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Yan Yan
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Current address: College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- Current address: Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| | - Iok In Christine Chio
- Institute for Cancer Genetics, Department of Genetics and Development and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Elisa de Stanchina
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, MSKCC
| | - Tuomas Tammela
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- David M. Rubenstein Center for Pancreatic Cancer, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
22
|
Tonelli C, Deschênes A, Gaeth V, Jensen A, Vithlani N, Yao MA, Zhao Z, Park Y, Tuveson DA. Ductal pancreatic cancer interception by FGFR2 abrogation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618726. [PMID: 39463990 PMCID: PMC11507947 DOI: 10.1101/2024.10.16.618726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Activating KRAS mutations are a key feature of pancreatic ductal adenocarcinoma (PDA) and drive tumor initiation and progression. However, mutant KRAS by itself is weakly oncogenic. The pathways that cooperate with mutant KRAS to induce tumorigenesis are less-defined. Analyzing organoids and murine and human pancreatic specimens, we found that the receptor tyrosine kinase FGFR2 was progressively up-regulated in mutant KRAS-driven metaplasia, pre-neoplasia and Classical PDA. Using genetic mouse models, we showed that FGFR2 supported mutant KRAS-driven transformation of acinar cells by promoting proliferation and MAPK pathway activation. FGFR2 abrogation significantly delayed tumor formation and extended the survival of these mice. Furthermore, we discovered that FGFR2 collaborated with EGFR and dual blockade of these receptor signaling pathways significantly reduced mutant KRAS-induced pre-neoplastic lesion formation. Together, our data have uncovered a pivotal role for FGFR2 in the early phases of pancreatic tumorigenesis, paving the way for future therapeutic applications of FGFR2 inhibitors for pancreatic cancer interception. STATEMENT OF SIGNIFICANCE Mutant KRAS-expressing pancreatic intraepithelial neoplasias (PanINs), the precursor lesions of PDA, are prevalent in the average healthy adult but rarely advance to invasive carcinoma. Here, we discovered that FGFR2 promoted PDA progression by amplifying mutant KRAS signaling and that inactivation of FGFR2 intercepted disease progression.
Collapse
|
23
|
Tamagawa H, Fujii M, Togasaki K, Seino T, Kawasaki S, Takano A, Toshimitsu K, Takahashi S, Ohta Y, Matano M, Kawasaki K, Machida Y, Sekine S, Machinaga A, Sasai K, Kodama Y, Kakiuchi N, Ogawa S, Hirano T, Seno H, Kitago M, Kitagawa Y, Iwasaki E, Kanai T, Sato T. Wnt-deficient and hypoxic environment orchestrates squamous reprogramming of human pancreatic ductal adenocarcinoma. Nat Cell Biol 2024; 26:1759-1772. [PMID: 39232216 DOI: 10.1038/s41556-024-01498-5] [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: 10/12/2022] [Accepted: 08/05/2024] [Indexed: 09/06/2024]
Abstract
Human pancreatic cancer is characterized by the molecular diversity encompassing native duct-like and squamous cell-like identities, but mechanisms underlying squamous transdifferentiation have remained elusive. To comprehensively capture the molecular diversity of human pancreatic cancer, we here profiled 65 patient-derived pancreatic cancer organoid lines, including six adenosquamous carcinoma lines. H3K27me3-mediated erasure of the ductal lineage specifiers and hijacking of the TP63-driven squamous-cell programme drove squamous-cell commitment, providing survival benefit in a Wnt-deficient environment and hypoxic conditions. Gene engineering of normal pancreatic duct organoids revealed that GATA6 loss and a Wnt-deficient environment, in concert with genetic or hypoxia-mediated inactivation of KDM6A, facilitate squamous reprogramming, which in turn enhances environmental fitness. EZH2 inhibition counterbalanced the epigenetic bias and curbed the growth of adenosquamous cancer organoids. Our results demonstrate how an adversarial microenvironment dictates the molecular and histological evolution of human pancreatic cancer and provide insights into the principles and significance of lineage conversion in human cancer.
Collapse
Affiliation(s)
- Hiroki Tamagawa
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| | - Kazuhiro Togasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Seino
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Shintaro Kawasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Ai Takano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Kohta Toshimitsu
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Kenta Kawasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Yujiro Machida
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Shigeki Sekine
- Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | | | | | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Nobuyuki Kakiuchi
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomonori Hirano
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Eisuke Iwasaki
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Kanai
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
24
|
Chen Q, Chen Z, Zhang J, Cai Y, Wu S, He D, Cheng K, Gu X, Cai Y, Wang X, Li Y, Zhang M, Wu Z, Peng B. Dual and triple gene combinations of KRT5, KRT17, and S100A2 identify basal-like subtype of pancreatic ductal adenocarcinoma and correlate with survival outcome. FASEB J 2024; 38:e23867. [PMID: 39101950 DOI: 10.1096/fj.202302484rr] [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: 12/01/2023] [Revised: 07/14/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024]
Abstract
There is a significant difference in prognosis and response to chemotherapy between basal and classical subtypes of pancreatic ductal adenocarcinoma (PDAC). Further biomarkers are required to identify subtypes of PDAC. We selected candidate biomarkers via review articles. Correlations between these candidate markers and the PDAC molecular subtype gene sets were analyzed using bioinformatics, confirming the biomarkers for identifying classical and basal subtypes. Subsequently, 298 PDAC patients were included, and their tumor tissues were immunohistochemically stratified using these biomarkers. Survival data underwent analysis, including Cox proportional hazards modeling. Our results indicate that the pairwise and triple combinations of KRT5/KRT17/S100A2 exhibit a higher correlation coefficient with the basal-like subtype gene set, whereas the corresponding combinations of GATA6/HNF4A/TFF1 show a higher correlation with the classical subtype gene set. Whether analyzing unmatched or propensity-matched data, the overall survival time was significantly shorter for the basal subtype compared with the classical subtype (p < .001), with basal subtype patients also facing a higher risk of mortality (HR = 4.017, 95% CI 2.675-6.032, p < .001). In conclusion, the combined expression of KRT5, KRT17, and S100A2, in both pairwise and triple combinations, independently predicts shorter overall survival in PDAC patients and likely identifies the basal subtype. Similarly, the combined expression of GATA6, HNF4A, and TFF1, in the same manner, may indicate the classical subtype. In our study, the combined application of established biomarkers offers valuable insights for the prognostic evaluation of PDAC patients.
Collapse
Affiliation(s)
- Qiangxing Chen
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
- West China School of Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Zixin Chen
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
- West China School of Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jing Zhang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
- West China School of Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Yunqiang Cai
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Shangdi Wu
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
- West China School of Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Du He
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ke Cheng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
- Division of Liver Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xiafei Gu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Cai
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
- Department of General Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xin Wang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yongbin Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
- Department of Minimal Invasive Surgery, Shangjin Nanfu Hospital, Chengdu, China
| | - Man Zhang
- Department of Minimal Invasive Surgery, Shangjin Nanfu Hospital, Chengdu, China
| | - Zhong Wu
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Bing Peng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Jiao Y, Lv Y, Liu M, Liu Y, Han M, Xiong X, Zhou H, Zhong J, Kang X, Su W. The modification role and tumor association with a methyltransferase: KMT2C. Front Immunol 2024; 15:1444923. [PMID: 39165358 PMCID: PMC11333232 DOI: 10.3389/fimmu.2024.1444923] [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: 06/06/2024] [Accepted: 07/22/2024] [Indexed: 08/22/2024] Open
Abstract
Histone methylation can affect chromosome structure and binding to other proteins, depending on the type of amino acid being modified and the number of methyl groups added, this modification may promote transcription of genes (H3K4me2, H3K4me3, and H3K79me3) or reduce transcription of genes (H3K9me2, H3K9me3, H3K27me2, H3K27me3, and H4K20me3). In addition, advances in tumor immunotherapy have shown that histone methylation as a type of protein post-translational modification is also involved in the proliferation, activation and metabolic reprogramming of immune cells in the tumor microenvironment. These post-translational modifications of proteins play a crucial role in regulating immune escape from tumors and immunotherapy. Lysine methyltransferases are important components of the post-translational histone methylation modification pathway. Lysine methyltransferase 2C (KMT2C), also known as MLL3, is a member of the lysine methyltransferase family, which mediates the methylation modification of histone 3 lysine 4 (H3K4), participates in the methylation of many histone proteins, and regulates a number of signaling pathways such as EMT, p53, Myc, DNA damage repair and other pathways. Studies of KMT2C have found that it is aberrantly expressed in many diseases, mainly tumors and hematological disorders. It can also inhibit the onset and progression of these diseases. Therefore, KMT2C may serve as a promising target for tumor immunotherapy for certain diseases. Here, we provide an overview of the structure of KMT2C, disease mechanisms, and diseases associated with KMT2C, and discuss related challenges.
Collapse
Affiliation(s)
- Yunjuan Jiao
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Yuanhao Lv
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Mingjie Liu
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yun Liu
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Miaomiao Han
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Xiwen Xiong
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Hongyan Zhou
- Xinxiang Key Laboratory of Precision Diagnosis and Treatment for Colorectal Cancer, Xinxiang First People’s Hospital, Xinxiang, China
| | - Jiateng Zhong
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
- Xinxiang Engineering Technology Research Center of Digestive Tumor Molecular Diagnosis, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xiaohong Kang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Wei Su
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Xinxiang Engineering Technology Research Center of Digestive Tumor Molecular Diagnosis, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
26
|
Felipe I, Real FX. PDAC heterogeneity resolved spatially at the single-cell level: new biological answers, new questions on optimal translation †. J Pathol 2024; 263:397-399. [PMID: 38828491 DOI: 10.1002/path.6293] [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: 03/23/2024] [Revised: 03/31/2024] [Accepted: 04/11/2024] [Indexed: 06/05/2024]
Abstract
Pancreatic cancer is a highly aggressive disease. Developing new strategies and using powerful methodologies for its early detection, coupled with in-depth comprehension of the mechanisms governing subtype evolution, will not only help to stratify PDAC patients' prognosis but also prevent unfavourable subtype plasticity upon treatment with chemotherapy. Michiels et al have developed a new approach to better capture PDAC heterogeneity at the single tumour duct spatial resolution level, leveraging detection of transcripts for mutant KRAS and multiple subtype markers. Their study sheds light on the association of mutant KRAS and PDAC phenotypic heterogeneity. The findings support functional cooperation of plastic tumour cells and opens new challenges towards PDAC patient stratification and therapeutic intervention. Pathology-based tools will be of prime importance to address these issues in a clinically meaningful manner. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Irene Felipe
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre - CNIO, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre - CNIO, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
- Departament de Medicina i Ciències de la Vida, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Besedina E, Supek F. Copy number losses of oncogenes and gains of tumor suppressor genes generate common driver mutations. Nat Commun 2024; 15:6139. [PMID: 39033140 PMCID: PMC11271286 DOI: 10.1038/s41467-024-50552-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 07/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cancer driver genes can undergo positive selection for various types of genetic alterations, including gain-of-function or loss-of-function mutations and copy number alterations (CNA). We investigated the landscape of different types of alterations affecting driver genes in 17,644 cancer exomes and genomes. We find that oncogenes may simultaneously exhibit signatures of positive selection and also negative selection in different gene segments, suggesting a method to identify additional tumor types where an oncogene is a driver or a vulnerability. Next, we characterize the landscape of CNA-dependent selection effects, revealing a general trend of increased positive selection on oncogene mutations not only upon CNA gains but also upon CNA deletions. Similarly, we observe a positive interaction between mutations and CNA gains in tumor suppressor genes. Thus, two-hit events involving point mutations and CNA are universally observed regardless of the type of CNA and may signal new therapeutic opportunities. An analysis with focus on the somatic CNA two-hit events can help identify additional driver genes relevant to a tumor type. By a global inference of point mutation and CNA selection signatures and interactions thereof across genes and tissues, we identify 9 evolutionary archetypes of driver genes, representing different mechanisms of (in)activation by genetic alterations.
Collapse
Affiliation(s)
- Elizaveta Besedina
- Institute for Research in Biomedicine (IRB Barcelona), 08028, Barcelona, Spain
| | - Fran Supek
- Institute for Research in Biomedicine (IRB Barcelona), 08028, Barcelona, Spain.
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen, Denmark.
- Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain.
| |
Collapse
|
30
|
Randriamanantsoa SJ, Raich MK, Saur D, Reichert M, Bausch AR. Coexisting mechanisms of luminogenesis in pancreatic cancer-derived organoids. iScience 2024; 27:110299. [PMID: 39055943 PMCID: PMC11269295 DOI: 10.1016/j.isci.2024.110299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/02/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
Lumens are crucial features of the tissue architecture in both the healthy exocrine pancreas, where ducts shuttle enzymes from the acini to the intestine, and in the precancerous lesions of the highly lethal pancreatic ductal adenocarcinoma (PDAC), similarly displaying lumens that can further develop into cyst-like structures. Branched pancreatic-cancer derived organoids capture key architectural features of both the healthy and diseased pancreas, including lumens. However, their transition from a solid mass of cells to a hollow tissue remains insufficiently explored. Here, we show that organoids display two orthogonal but complementary lumen formation mechanisms: one relying on fluid intake for multiple microlumen nucleation, swelling and fusion, and the other involving the death of a central cell population, thereby hollowing out cavities. These results shed further light on the processes of luminogenesis, deepening our understanding of the early formation of PDAC precancerous lesions, including cystic neoplasia.
Collapse
Affiliation(s)
- Samuel J. Randriamanantsoa
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Chair for Cellular Biophysics E27, 85748 Garching, Germany
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
| | - Marion K. Raich
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Chair for Cellular Biophysics E27, 85748 Garching, Germany
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
| | - Dieter Saur
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Medical Clinic and Polyclinic II, 81675 Munich, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner site Munich, 69120 Heidelberg, Germany
| | - Maximilian Reichert
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Medical Clinic and Polyclinic II, 81675 Munich, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner site Munich, 69120 Heidelberg, Germany
- Technical University of Munich, Klinikum rechts der Isar, Medical Clinic and Polyclinic II, Translational Pancreatic Cancer Research Center, 81675 Munich, Germany
| | - Andreas R. Bausch
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Chair for Cellular Biophysics E27, 85748 Garching, Germany
- Technical University of Munich, Center for Functional Protein Assemblies (CPA), 85748 Garching, Germany
- Technical University of Munich, Center for Organoid Systems and Tissue Engineering (COS), 85748 Garching, Germany
| |
Collapse
|
31
|
Maurer HC, Garcia-Curiel A, Holmstrom SR, Castillo C, Palermo CF, Sastra SA, Andren A, Zhang L, Le Large TYS, Sagalovskiy I, Ross DR, Wong W, Shaw K, Genkinger J, Manji GA, Iuga AC, Schmid RM, Johnson K, Badgley MA, Lyssiotis CA, Shah Y, Califano A, Olive KP. Ras-dependent activation of BMAL2 regulates hypoxic metabolism in pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.19.533333. [PMID: 36993718 PMCID: PMC10055246 DOI: 10.1101/2023.03.19.533333] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
To identify drivers of malignancy in human pancreatic ductal adenocarcinoma (PDAC), we performed regulatory network analysis on a large collection of expression profiles from laser capture microdissected samples of PDAC and benign precursors. We discovered that BMAL2 plays a role in the initiation, progression, post resection survival, and KRAS activity in PDAC. Functional analysis of BMAL2 target genes led us to hypothesize that it plays a role in regulating the response to hypoxia, a critical but poorly understood feature of PDAC physiology. Knockout of BMAL2 in multiple human PDAC cell lines revealed effects on viability and invasion, particularly under hypoxic conditions. Loss of BMAL2 also affected glycolysis and other metabolic processes. We found that BMAL2 directly regulates hypoxia-responsive target genes. We also found that BMAL2 is necessary for the stabilization of HIF1A upon exposure to hypoxia, but destabilizes HIF2A under hypoxia. These data demonstrate that BMAL2 is a master transcriptional regulator of hypoxia responses in PDAC and may serve as a long-sought molecular switch that distinguishes HIF1A- and HIF2A-dependent modes of hypoxic metabolism.
Collapse
Affiliation(s)
- H Carlo Maurer
- Department of Internal Medicine II, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Alvaro Garcia-Curiel
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Columbia University Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY
| | - Sam R Holmstrom
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Cristina Castillo
- Department of Molecular & Integrative Physiology and Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI
| | - Carmine F Palermo
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Columbia University Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY
| | - Steven A Sastra
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Columbia University Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY
| | - Anthony Andren
- Department of Molecular & Integrative Physiology and Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI
| | - Li Zhang
- Department of Molecular & Integrative Physiology and Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI
| | - Tessa Y S Le Large
- Department of Surgery, Amsterdam UMC, Location Vrije Universiteit, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Irina Sagalovskiy
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Daniel R Ross
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Columbia University Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY
| | - Winston Wong
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Kaitlin Shaw
- Division of GI/Endocrine Surgery, Department of Surgery, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Jeanine Genkinger
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY
| | - Gulam A Manji
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Alina C Iuga
- Department of Pathology, Columbia University Irving Medical Center, New York, NY
| | - Roland M Schmid
- Department of Internal Medicine II, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | | | - Michael A Badgley
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Columbia University Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY
| | - Costas A Lyssiotis
- Department of Molecular & Integrative Physiology and Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI
| | - Yatrik Shah
- Department of Molecular & Integrative Physiology and Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI
| | - Andrea Califano
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Darwin Therapeutics, New York, NY
- Chan Zuckerberg Biohub, New York, NY
| | - Kenneth P Olive
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Columbia University Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
32
|
Qin X, Gan Z, Liu H, Tao T, He J, Li X, Shang D, Li X, Xie F, Qin J. A Pump-Free Strategy for the Controllable Generation of Alginate Microgels as Cellular Microcarriers. ACS Biomater Sci Eng 2024; 10:3958-3967. [PMID: 38711418 DOI: 10.1021/acsbiomaterials.4c00375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Microgels are advanced scaffolds for tissue engineering due to their proper biodegradability, good biocompatibility, and high specific surface area for effective oxygen and nutrient transfer. However, most of the current monodispersed microgel fabrication systems rely heavily on various precision pumps, which highly increase the cost and complexity of their downstream application. In this work, we developed a simple and facile system for the controllable generation of uniform alginate microgels by integrating a gas-shearing strategy into a glass microfluidic device. Importantly, the cell-laden microgels can be rapidly prepared in a pump-free manner under an all-aqueous environment. The three-dimensional cultured green fluorescent protein-human A549 cells in alginate microgels exhibited enhanced stemness and drug resistance compared to those under two-dimensional conditions. The pancreatic cancer organoids in alginate microgels exhibited some of the key features of pancreatic cancer. The proposed microgels showed decent monodispersity, biocompatibility, and versatility, providing great opportunities in various biomedical applications such as microcarrier fabricating, organoid engineering, and high-throughput drug screening.
Collapse
Affiliation(s)
- Xinyuan Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Science, Beijing 100049, China
| | - Zhongqiao Gan
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Science, Beijing 100049, China
| | - Haitao Liu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Tingting Tao
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jia He
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Xianliang Li
- Department of HBP Surgery, Beijing Chao Yang Hospital, the Capital Medical University, Beijing 100020, China
| | - Dong Shang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, No.222 Zhongshan Road, Dalian 116011, China
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, No.222 Zhongshan Road, Dalian 116011, China
| | - Xiang Li
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, China
| | - Fuwei Xie
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Science, Beijing 100049, China
- Beijing Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China
- University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| |
Collapse
|
33
|
Tomić G, Sheridan C, Refermat AY, Baggelaar MP, Sipthorp J, Sudarshan B, Ocasio CA, Suárez-Bonnet A, Priestnall SL, Herbert E, Tate EW, Downward J. Palmitoyl transferase ZDHHC20 promotes pancreatic cancer metastasis. Cell Rep 2024; 43:114224. [PMID: 38733589 DOI: 10.1016/j.celrep.2024.114224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/04/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Metastasis is one of the defining features of pancreatic ductal adenocarcinoma (PDAC) that contributes to poor prognosis. In this study, the palmitoyl transferase ZDHHC20 was identified in an in vivo short hairpin RNA (shRNA) screen as critical for metastatic outgrowth, with no effect on proliferation and migration in vitro or primary PDAC growth in mice. This phenotype is abrogated in immunocompromised animals and animals with depleted natural killer (NK) cells, indicating that ZDHHC20 affects the interaction of tumor cells and the innate immune system. Using a chemical genetics platform for ZDHHC20-specific substrate profiling, a number of substrates of this enzyme were identified. These results describe a role for palmitoylation in enabling distant metastasis that could not have been detected using in vitro screening approaches and identify potential effectors through which ZDHHC20 promotes metastasis of PDAC.
Collapse
Affiliation(s)
- Goran Tomić
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Clare Sheridan
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | | | - Marc P Baggelaar
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Imperial College London, Department of Chemistry, Molecular Sciences Research Hub, 80 Wood Lane, London W12 0BZ, UK
| | - James Sipthorp
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Imperial College London, Department of Chemistry, Molecular Sciences Research Hub, 80 Wood Lane, London W12 0BZ, UK
| | | | - Cory A Ocasio
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Alejandro Suárez-Bonnet
- The Royal Veterinary College, Department of Pathobiology & Population Sciences, Hawkshead Lane, Hatfield AL9 7TA, UK; Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Simon L Priestnall
- The Royal Veterinary College, Department of Pathobiology & Population Sciences, Hawkshead Lane, Hatfield AL9 7TA, UK; Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Eleanor Herbert
- The Royal Veterinary College, Department of Pathobiology & Population Sciences, Hawkshead Lane, Hatfield AL9 7TA, UK; Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Edward W Tate
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Imperial College London, Department of Chemistry, Molecular Sciences Research Hub, 80 Wood Lane, London W12 0BZ, UK
| | - Julian Downward
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
34
|
Seeger N, Gutknecht S, Zschokke I, Fleischmann I, Roth N, Metzger J, Weber M, Breitenstein S, Grochola LF. A Predictive Noninvasive Single-Nucleotide Variation-Based Biomarker Signature for Resectable Pancreatic Cancer: Protocol for a Prospective Validation Study. JMIR Res Protoc 2024; 13:e54042. [PMID: 38635586 PMCID: PMC11130767 DOI: 10.2196/54042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Single-nucleotide variations (SNVs; formerly SNPs) are inherited genetic variants that can be easily determined in routine clinical practice using a simple blood or saliva test. SNVs have potential to serve as noninvasive biomarkers for predicting cancer-specific patient outcomes after resection of pancreatic ductal adenocarcinoma (PDAC). Two recent analyses led to the identification and validation of three SNVs in the CD44 and CHI3L2 genes (rs187115, rs353630, and rs684559), which can be used as predictive biomarkers to help select patients most likely to benefit from pancreatic resection. These variants were associated with an over 2-fold increased risk for tumor-related death in three independent PDAC study cohorts from Europe and the United States, including The Cancer Genome Atlas cohorts (reaching a P value of 1×10-8). However, these analyses were limited by the inherent biases of a retrospective study design, such as selection and publication biases, thereby limiting the clinical use of these promising biomarkers in guiding PDAC therapy. OBJECTIVE To overcome the limitations of previous retrospectively designed studies and translate the findings into clinical practice, we aim to validate the association of the identified SNVs with survival in a controlled setting using a prospective cohort of patients with PDAC following pancreatic resection. METHODS All patients with PDAC who will undergo pancreatic resection at three participating hospitals in Switzerland and fulfill the inclusion criteria will be included in the study consecutively. The SNV genotypes will be determined using standard genotyping techniques from patient blood samples. For each genotyped locus, log-rank and Cox multivariate regression tests will be performed, accounting for the relevant covariates American Joint Committee on Cancer stage and resection status. Clinical follow-up data will be collected for at least 3 years. Sample size calculation resulted in a required sample of 150 patients to sufficiently power the analysis. RESULTS The follow-up data collection started in August 2019 and the estimated end of data collection will be in May 2027. The study is still recruiting participants and 142 patients have been recruited as of November 2023. The DNA extraction and genotyping of the SNVs will be performed after inclusion of the last patient. Since no SNV genotypes have been determined, no data analysis has been performed to date. The results are expected to be published in 2027. CONCLUSIONS This is the first prospective study of the CD44 and CHI3L2 SNV-based biomarker signature in PDAC. A prospective validation of this signature would enable its clinical use as a noninvasive predictive biomarker of survival after pancreatic resection that is readily available at the time of diagnosis and can assist in guiding PDAC therapy. The results of this study may help to individualize treatment decisions and potentially improve patient outcomes. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/54042.
Collapse
Affiliation(s)
- Nico Seeger
- Department of Visceral and Thoracic Surgery, Cantonal Hospital of Winterthur, Winterthur, Switzerland
| | - Stefan Gutknecht
- Department of Visceral, Thoracic and Cardiovascular Surgery, Triemli Hospital, Zurich, Switzerland
| | - Irin Zschokke
- Department of General and Visceral Surgery, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Isabella Fleischmann
- Department of General and Visceral Surgery, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Nadja Roth
- Department of General and Visceral Surgery, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Jürg Metzger
- Department of General and Visceral Surgery, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Markus Weber
- Department of Visceral, Thoracic and Cardiovascular Surgery, Triemli Hospital, Zurich, Switzerland
| | - Stefan Breitenstein
- Department of Visceral and Thoracic Surgery, Cantonal Hospital of Winterthur, Winterthur, Switzerland
| | - Lukasz Filip Grochola
- Department of Visceral and Thoracic Surgery, Cantonal Hospital of Winterthur, Winterthur, Switzerland
| |
Collapse
|
35
|
Park JK, Jeong HO, Kim H, Choi JH, Lee EM, Kim S, Jang J, Choi DWY, Lee SH, Kim KM, Jang KT, Lee KH, Lee KT, Lee MW, Lee JK, Lee S. Single-cell transcriptome analysis reveals subtype-specific clonal evolution and microenvironmental changes in liver metastasis of pancreatic adenocarcinoma and their clinical implications. Mol Cancer 2024; 23:87. [PMID: 38702773 PMCID: PMC11067162 DOI: 10.1186/s12943-024-02003-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/16/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Intratumoral heterogeneity (ITH) and tumor microenvironment (TME) of pancreatic ductal adenocarcinoma (PDAC) play important roles in tumor evolution and patient outcomes. However, the precise characterization of diverse cell populations and their crosstalk associated with PDAC progression and metastasis is still challenging. METHODS We performed single-cell RNA sequencing (scRNA-seq) of treatment-naïve primary PDAC samples with and without paired liver metastasis samples to understand the interplay between ITH and TME in the PDAC evolution and its clinical associations. RESULTS scRNA-seq analysis revealed that even a small proportion (22%) of basal-like malignant ductal cells could lead to poor chemotherapy response and patient survival and that epithelial-mesenchymal transition programs were largely subtype-specific. The clonal homogeneity significantly increased with more prevalent and pronounced copy number gains of oncogenes, such as KRAS and ETV1, and losses of tumor suppressor genes, such as SMAD2 and MAP2K4, along PDAC progression and metastasis. Moreover, diverse immune cell populations, including naïve SELLhi regulatory T cells (Tregs) and activated TIGIThi Tregs, contributed to shaping immunosuppressive TMEs of PDAC through cellular interactions with malignant ductal cells in PDAC evolution. Importantly, the proportion of basal-like ductal cells negatively correlated with that of immunoreactive cell populations, such as cytotoxic T cells, but positively correlated with that of immunosuppressive cell populations, such as Tregs. CONCLUSION We uncover that the proportion of basal-like subtype is a key determinant for chemotherapy response and patient outcome, and that PDAC clonally evolves with subtype-specific dosage changes of cancer-associated genes by forming immunosuppressive microenvironments in its progression and metastasis.
Collapse
Affiliation(s)
- Joo Kyung Park
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Hyoung-Oh Jeong
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Hyemin Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Ho Choi
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun Mi Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seunghoon Kim
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Jinho Jang
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - David Whee-Young Choi
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Se-Hoon Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyoung Mee Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kee-Taek Jang
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kwang Hyuck Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyu Taek Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Min Woo Lee
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Jong Kyun Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Semin Lee
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea.
| |
Collapse
|
36
|
Nguyen A, Mustafa AHM, Leydecker AK, Halilovic M, Murr J, Butter F, Krämer OH. The protein phosphatase-2A subunit PR130 is involved in the formation of cytotoxic protein aggregates in pancreatic ductal adenocarcinoma cells. Cell Commun Signal 2024; 22:217. [PMID: 38570831 PMCID: PMC10993613 DOI: 10.1186/s12964-024-01597-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
As a major source of cellular serine and threonine phosphatase activity, protein phosphatase-2A (PP2A) modulates signaling pathways in health and disease. PP2A complexes consist of catalytic, scaffolding, and B-type subunits. Seventeen PP2A B-type subunits direct PP2A complexes to selected substrates. It is ill-defined how PP2A B-type subunits determine the growth and drug responsiveness of tumor cells. Pancreatic ductal adenocarcinoma (PDAC) is a disease with poor prognosis. We analyzed the responses of murine and human mesenchymal and epithelial PDAC cells to the specific PP2A inhibitor phendione. We assessed protein levels by immunoblot and proteomics and cell fate by flow cytometry, confocal microscopy, and genetic manipulation. We show that murine mesenchymal PDAC cells express significantly higher levels of the PP2A B-type subunit PR130 than epithelial PDAC cells. This overexpression of PR130 is associated with a dependency of such metastasis-prone cells on the catalytic activity of PP2A. Phendione induces apoptosis and an accumulation of cytotoxic protein aggregates in murine mesenchymal and human PDAC cells. These processes occur independently of the frequently mutated tumor suppressor p53. Proteomic analyses reveal that phendione upregulates the chaperone HSP70 in mesenchymal PDAC cells. Inhibition of HSP70 promotes phendione-induced apoptosis and phendione promotes a proteasomal degradation of PR130. Genetic elimination of PR130 sensitizes murine and human PDAC cells to phendione-induced apoptosis and protein aggregate formation. These data suggest that the PP2A-PR130 complex dephosphorylates and thereby prevents the aggregation of proteins in tumor cells.
Collapse
Affiliation(s)
- Alexandra Nguyen
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher St. 67, 55131, Mainz, Germany
| | - Al-Hassan M Mustafa
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher St. 67, 55131, Mainz, Germany
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
| | - Alessa K Leydecker
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher St. 67, 55131, Mainz, Germany
| | - Melisa Halilovic
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher St. 67, 55131, Mainz, Germany
| | - Janine Murr
- Medical Clinic and Polyclinic II, Klinikum Rechts Der Isar, Technical University Munich, 81675, Munich, Germany
| | - Falk Butter
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128, Mainz, Germany
- Federal Research Institute for Animal Health, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Oliver H Krämer
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher St. 67, 55131, Mainz, Germany.
| |
Collapse
|
37
|
Guo X, Bian X, Li Y, Zhu X, Zhou X. The intricate dance of tumor evolution: Exploring immune escape, tumor migration, drug resistance, and treatment strategies. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167098. [PMID: 38412927 DOI: 10.1016/j.bbadis.2024.167098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/14/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024]
Abstract
Recent research has unveiled fascinating insights into the intricate mechanisms governing tumor evolution. These studies have illuminated how tumors adapt and proliferate by exploiting various factors, including immune evasion, resistance to therapeutic drugs, genetic mutations, and their ability to adapt to different environments. Furthermore, investigations into tumor heterogeneity and chromosomal aberrations have revealed the profound complexity that underlies the evolution of cancer. Emerging findings have also underscored the role of viral influences in the development and progression of cancer, introducing an additional layer of complexity to the field of oncology. Tumor evolution is a dynamic and complex process influenced by various factors, including immune evasion, drug resistance, tumor heterogeneity, and viral influences. Understanding these elements is indispensable for developing more effective treatments and advancing cancer therapies. A holistic approach to studying and addressing tumor evolution is crucial in the ongoing battle against cancer. The main goal of this comprehensive review is to explore the intricate relationship between tumor evolution and critical aspects of cancer biology. By delving into this complex interplay, we aim to provide a profound understanding of how tumors evolve, adapt, and respond to treatment strategies. This review underscores the pivotal importance of comprehending tumor evolution in shaping effective approaches to cancer treatment.
Collapse
Affiliation(s)
- Xiaojun Guo
- Department of Immunology, School of Medicine, Nantong University, Nantong, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xiaonan Bian
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Yitong Li
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China.
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China.
| |
Collapse
|
38
|
Linehan A, O’Reilly M, McDermott R, O’Kane GM. Targeting KRAS mutations in pancreatic cancer: opportunities for future strategies. Front Med (Lausanne) 2024; 11:1369136. [PMID: 38576709 PMCID: PMC10991798 DOI: 10.3389/fmed.2024.1369136] [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: 01/11/2024] [Accepted: 02/22/2024] [Indexed: 04/06/2024] Open
Abstract
Targeting the RAS pathway remains the holy grail of precision oncology. In the case of pancreatic ductal adenocarcinomas (PDAC), 90-92% harbor mutations in the oncogene KRAS, triggering canonical MAPK signaling. The smooth structure of the altered KRAS protein without a binding pocket and its affinity for GTP have, in the past, hampered drug development. The emergence of KRASG12C covalent inhibitors has provided renewed enthusiasm for targeting KRAS. The numerous pathways implicated in RAS activation do, however, lead to the development of early resistance. In addition, the dense stromal niche and immunosuppressive microenvironment dictated by oncogenic KRAS can influence treatment responses, highlighting the need for a combination-based approach. Given that mutations in KRAS occur early in PDAC tumorigenesis, an understanding of its pleiotropic effects is key to progress in this disease. Herein, we review current perspectives on targeting KRAS with a focus on PDAC.
Collapse
Affiliation(s)
- Anna Linehan
- Department of Medical Oncology, St Vincent’s University Hospital, Dublin, Ireland
| | - Mary O’Reilly
- Department of Medical Oncology, St Vincent’s University Hospital, Dublin, Ireland
| | - Ray McDermott
- Department of Medical Oncology, St Vincent’s University Hospital, Dublin, Ireland
| | - Grainne M. O’Kane
- Department of Medical Oncology, St James’s Hospital, Dublin, Ireland
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
39
|
Tsanov KM, Barriga FM, Ho YJ, Alonso-Curbelo D, Livshits G, Koche RP, Baslan T, Simon J, Tian S, Wuest AN, Luan W, Wilkinson JE, Masilionis I, Dimitrova N, Iacobuzio-Donahue CA, Chaligné R, Pe’er D, Massagué J, Lowe SW. Metastatic site influences driver gene function in pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.17.585402. [PMID: 38562717 PMCID: PMC10983983 DOI: 10.1101/2024.03.17.585402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Driver gene mutations can increase the metastatic potential of the primary tumor1-3, but their role in sustaining tumor growth at metastatic sites is poorly understood. A paradigm of such mutations is inactivation of SMAD4 - a transcriptional effector of TGFβ signaling - which is a hallmark of multiple gastrointestinal malignancies4,5. SMAD4 inactivation mediates TGFβ's remarkable anti- to pro-tumorigenic switch during cancer progression and can thus influence both tumor initiation and metastasis6-14. To determine whether metastatic tumors remain dependent on SMAD4 inactivation, we developed a mouse model of pancreatic ductal adenocarcinoma (PDAC) that enables Smad4 depletion in the pre-malignant pancreas and subsequent Smad4 reactivation in established metastases. As expected, Smad4 inactivation facilitated the formation of primary tumors that eventually colonized the liver and lungs. By contrast, Smad4 reactivation in metastatic disease had strikingly opposite effects depending on the tumor's organ of residence: suppression of liver metastases and promotion of lung metastases. Integrative multiomic analysis revealed organ-specific differences in the tumor cells' epigenomic state, whereby the liver and lungs harbored chromatin programs respectively dominated by the KLF and RUNX developmental transcription factors, with Klf4 depletion being sufficient to reverse Smad4's tumor-suppressive activity in liver metastases. Our results show how epigenetic states favored by the organ of residence can influence the function of driver genes in metastatic tumors. This organ-specific gene-chromatin interplay invites consideration of anatomical site in the interpretation of tumor genetics, with implications for the therapeutic targeting of metastatic disease.
Collapse
Affiliation(s)
- Kaloyan M. Tsanov
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francisco M. Barriga
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Yu-Jui Ho
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Direna Alonso-Curbelo
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Geulah Livshits
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard P. Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timour Baslan
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Biomedical Sciences, School of Veterinary Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - Janelle Simon
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sha Tian
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexandra N. Wuest
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wei Luan
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John E. Wilkinson
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Ignas Masilionis
- Computational & Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nevenka Dimitrova
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christine A. Iacobuzio-Donahue
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronan Chaligné
- Computational & Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dana Pe’er
- Computational & Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Joan Massagué
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Scott W. Lowe
- Cancer Biology & Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
40
|
Ho IL, Li CY, Wang F, Zhao L, Liu J, Yen EY, Dyke CA, Shah R, Liu Z, Çetin AO, Chu Y, Citron F, Attanasio S, Corti D, Darbaniyan F, Del Poggetto E, Loponte S, Liu J, Soeung M, Chen Z, Jiang S, Jiang H, Inoue A, Gao S, Deem A, Feng N, Ying H, Kim M, Giuliani V, Genovese G, Zhang J, Futreal A, Maitra A, Heffernan T, Wang L, Do KA, Gargiulo G, Draetta G, Carugo A, Lin R, Viale A. Clonal dominance defines metastatic dissemination in pancreatic cancer. SCIENCE ADVANCES 2024; 10:eadd9342. [PMID: 38478609 DOI: 10.1126/sciadv.add9342] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 02/08/2024] [Indexed: 02/08/2025]
Abstract
Tumors represent ecosystems where subclones compete during tumor growth. While extensively investigated, a comprehensive picture of the interplay of clonal lineages during dissemination is still lacking. Using patient-derived pancreatic cancer cells, we created orthotopically implanted clonal replica tumors to trace clonal dynamics of unperturbed tumor expansion and dissemination. This model revealed the multifaceted nature of tumor growth, with rapid changes in clonal fitness leading to continuous reshuffling of tumor architecture and alternating clonal dominance as a distinct feature of cancer growth. Regarding dissemination, a large fraction of tumor lineages could be found at secondary sites each having distinctive organ growth patterns as well as numerous undescribed behaviors such as abortive colonization. Paired analysis of primary and secondary sites revealed fitness as major contributor to dissemination. From the analysis of pro- and nonmetastatic isogenic subclones, we identified a transcriptomic signature able to identify metastatic cells in human tumors and predict patients' survival.
Collapse
Affiliation(s)
- I-Lin Ho
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chieh-Yuan Li
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fuchenchu Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jingjing Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Er-Yen Yen
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Charles A Dyke
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rutvi Shah
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhaoliang Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ali Osman Çetin
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Yanshuo Chu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francesca Citron
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sergio Attanasio
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Denise Corti
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Faezeh Darbaniyan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edoardo Del Poggetto
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sara Loponte
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jintan Liu
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melinda Soeung
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ziheng Chen
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shan Jiang
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hong Jiang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Akira Inoue
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sisi Gao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Angela Deem
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ningping Feng
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haoqiang Ying
- Department of Cellular and Molecular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Virginia Giuliani
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy Heffernan
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gaetano Gargiulo
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Giulio Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessandro Carugo
- TRACTION platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
41
|
Choi MH, Yoon SB, Lee YJ, Jung ES, Pak S, Han D, Nickel D. Rim enhancement of pancreatic ductal adenocarcinoma: investigating the relationship with DCE-MRI-based radiomics and next-generation sequencing. Front Oncol 2024; 14:1304187. [PMID: 38525415 PMCID: PMC10959187 DOI: 10.3389/fonc.2024.1304187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/16/2024] [Indexed: 03/26/2024] Open
Abstract
PURPOSE To identify the clinical and genetic variables associated with rim enhancement of pancreatic ductal adenocarcinoma (PDAC) and to develop a dynamic contrast-enhanced (DCE) MRI-based radiomics model for predicting the genetic status from next-generation sequencing (NGS). MATERIALS AND METHODS Patients with PDAC, who underwent pretreatment pancreatic DCE-MRI between November 2019 and July 2021, were eligible in this prospective study. Two radiologists evaluated presence of rim enhancement in PDAC, a known radiological prognostic indicator, on DCE MRI. NGS was conducted for the tissue from the lesion. The Mann-Whitney U and Chi-square tests were employed to identify clinical and genetic variables associated with rim enhancement in PDAC. For continuous variables predicting rim enhancement, the cutoff value was set based on the Youden's index from the receiver operating characteristic (ROC) curve. Radiomics features were extracted from a volume-of-interest of PDAC on four DCE maps (Ktrans, Kep, Ve, and iAUC). A random forest (RF) model was constructed using 10 selected radiomics features from a pool of 392 original features. This model aimed to predict the status of significant NGS variables associated with rim enhancement. The performance of the model was validated using test set. RESULTS A total of 55 patients (32 men; median age 71 years) were randomly assigned to the training (n = 41) and test (n = 14) sets. In the training set, KRAS, TP53, CDKN2A, and SMAD4 mutation rates were 92.3%, 61.8%, 14.5%, and 9.1%, respectively. Tumor size and KRAS variant allele frequency (VAF) differed between rim-enhancing (n = 12) and nonrim-enhancing (n = 29) PDACs with a cutoff of 17.22%. The RF model's average AUC from 10-fold cross-validation for predicting KRAS VAF status was 0.698. In the test set comprising 6 tumors with low KRAS VAF and 8 with high KRAS VAF, the RF model's AUC reached 1.000, achieving a sensitivity of 75.0%, specificity of 100% and accuracy of 87.5%. CONCLUSION Rim enhancement of PDAC is associated with KRAS VAF derived from NGS-based genetic information. For predicting the KRAS VAF status in PDAC, a radiomics model based on DCE maps showed promising results.
Collapse
Affiliation(s)
- Moon Hyung Choi
- Department of Radiology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Bae Yoon
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Young Joon Lee
- Department of Radiology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sun Jung
- Department of Hospital Pathology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seongyong Pak
- Research Collaboration, Siemens Healthineers Ltd., Seoul, Republic of Korea
| | - Dongyeob Han
- Research Collaboration, Siemens Healthineers Ltd., Seoul, Republic of Korea
| | - Dominik Nickel
- MR Applications Predevelopment, Siemens Healthcare GmbH, Erlangen, Germany
| |
Collapse
|
42
|
Zheng C, Wang J, Wang J, Zhang Q, Liang T. Cell of Origin of Pancreatic cancer: Novel Findings and Current Understanding. Pancreas 2024; 53:e288-e297. [PMID: 38277420 PMCID: PMC11882172 DOI: 10.1097/mpa.0000000000002301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/08/2023] [Indexed: 01/28/2024]
Abstract
ABSTRACT Pancreatic ductal adenocarcinoma (PDAC) stands as one of the most lethal diseases globally, boasting a grim 5-year survival prognosis. The origin cell and the molecular signaling pathways that drive PDAC progression are not entirely understood. This review comprehensively outlines the categorization of PDAC and its precursor lesions, expounds on the creation and utility of genetically engineered mouse models used in PDAC research, compiles a roster of commonly used markers for pancreatic progenitors, duct cells, and acinar cells, and briefly addresses the mechanisms involved in the progression of PDAC. We acknowledge the value of precise markers and suitable tracing tools to discern the cell of origin, as it can facilitate the creation of more effective models for PDAC exploration. These conclusions shed light on our existing understanding of foundational genetically engineered mouse models and focus on the origin and development of PDAC.
Collapse
Affiliation(s)
- Chenlei Zheng
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
| | - Jianing Wang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
| | - Junli Wang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
| | - Qi Zhang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province
- Zhejiang University Cancer Center, Hangzhou, China
| | - Tingbo Liang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province
- Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
43
|
Huang YK, Cheng WC, Kuo TT, Yang JC, Wu YC, Wu HH, Lo CC, Hsieh CY, Wong SC, Lu CH, Wu WL, Liu SJ, Li YC, Lin CC, Shen CN, Hung MC, Lin JT, Yeh CC, Sher YP. Inhibition of ADAM9 promotes the selective degradation of KRAS and sensitizes pancreatic cancers to chemotherapy. NATURE CANCER 2024; 5:400-419. [PMID: 38267627 DOI: 10.1038/s43018-023-00720-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 12/19/2023] [Indexed: 01/26/2024]
Abstract
Kirsten rat sarcoma virus (KRAS) signaling drives pancreatic ductal adenocarcinoma (PDAC) malignancy, which is an unmet clinical need. Here, we identify a disintegrin and metalloproteinase domain (ADAM)9 as a modulator of PDAC progression via stabilization of wild-type and mutant KRAS proteins. Mechanistically, ADAM9 loss increases the interaction of KRAS with plasminogen activator inhibitor 1 (PAI-1), which functions as a selective autophagy receptor in conjunction with light chain 3 (LC3), triggering lysosomal degradation of KRAS. Suppression of ADAM9 by a small-molecule inhibitor restricts disease progression in spontaneous models, and combination with gemcitabine elicits dramatic regression of patient-derived tumors. Our findings provide a promising strategy to target the KRAS signaling cascade and demonstrate a potential modality to enhance sensitivity to chemotherapy in PDAC.
Collapse
Affiliation(s)
- Yu-Kai Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Wei-Chung Cheng
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan
| | - Ting-Ting Kuo
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Juan-Cheng Yang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yang-Chang Wu
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Heng-Hsiung Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chia-Chien Lo
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Ying Hsieh
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Sze-Ching Wong
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chih-Hao Lu
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wan-Ling Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Jen Liu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Chuan Li
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Ching-Chan Lin
- Division of Hematology and Oncology, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Ning Shen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Mien-Chie Hung
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jaw-Town Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan
| | - Chun-Chieh Yeh
- Department of Medicine, School of Medicine, China Medical University, Taichung, Taiwan.
- Department of Surgery, Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan.
| | - Yuh-Pyng Sher
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan.
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan.
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan.
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, Taiwan.
| |
Collapse
|
44
|
Francescone R, Crawford HC, Vendramini-Costa DB. Rethinking the Roles of Cancer-Associated Fibroblasts in Pancreatic Cancer. Cell Mol Gastroenterol Hepatol 2024; 17:737-743. [PMID: 38316215 PMCID: PMC10966284 DOI: 10.1016/j.jcmgh.2024.01.022] [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: 11/19/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Bearing a dismal 5-year survival rate, pancreatic ductal adenocarcinoma (PDAC) is a challenging disease that features a unique fibroinflammatory tumor microenvironment. As major components of the PDAC tumor microenvironment, cancer-associated fibroblasts are still poorly understood and their contribution to the several hallmarks of PDAC, such as resistance to therapies, immunosuppression, and high incidence of metastasis, is likely underestimated. There have been encouraging advances in the understanding of these fascinating cells, but many controversies remain, leaving the field still actively exploring the full scope of their contributions in PDAC progression. Here we pose several important considerations regarding PDAC cancer-associated fibroblast functions. We posit that transcriptomic analyses be interpreted with caution, when aiming to uncover the functional contributions of these cells. Moreover, we propose that normalizing these functions, rather than eliminating them, will provide the opportunity to enhance therapeutic response. Finally, we propose that cancer-associated fibroblasts should not be studied in isolation, but in conjunction with its extracellular matrix, because their respective functions are coordinated and concordant.
Collapse
Affiliation(s)
- Ralph Francescone
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Howard C Crawford
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Debora Barbosa Vendramini-Costa
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan.
| |
Collapse
|
45
|
Kern J, Schilling D, Schneeweis C, Schmid RM, Schneider G, Combs SE, Dobiasch S. Identification of the unfolded protein response pathway as target for radiosensitization in pancreatic cancer. Radiother Oncol 2024; 191:110059. [PMID: 38135186 DOI: 10.1016/j.radonc.2023.110059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND AND PURPOSE Due to the high intrinsic radioresistance of pancreatic ductal adenocarcinoma (PDAC), radiotherapy (RT) is only beneficial in 30% of patients. Therefore, this study aimed to identify targets to improve the efficacy of RT in PDAC. MATERIALS AND METHODS Alamar Blue proliferation and colony formation assay (CFA) were used to determine the radioresponse of a cohort of 38 murine PDAC cell lines. A gene set enrichment analysis was performed to reveal differentially expressed pathways. CFA, cell cycle distribution, γH2AX FACS analysis, and Caspase 3/7 SYTOX assay were used to examine the effect of a combination treatment using KIRA8 as an IRE1α-inhibitor and Ceapin-A7 as an inhibitor against ATF6. RESULTS The unfolded protein response (UPR) was identified as a pathway highly expressed in radioresistant cell lines. Using the IRE1α-inhibitor KIRA8 or the ATF6-inhibitor Ceapin-A7 in combination with radiation, a radiosensitizing effect was observed in radioresistant cell lines, but no substantial alteration of the radioresponse in radiosensitive cell lines. Mechanistically, increased apoptosis by KIRA8 in combination with radiation and a cell cycle arrest in the G1 phase after ATF6 inhibition and radiation have been observed in radioresistant cell lines. CONCLUSION So, our data show evidence that the UPR is involved in radioresistance of PDAC. Increased apoptosis and a G1 cell cycle arrest seem to be responsible for the radiosensitizing effect of UPR inhibition. These findings are supportive for developing novel combination treatment concepts in PDAC to overcome radioresistance.
Collapse
Affiliation(s)
- Jana Kern
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany
| | - Daniela Schilling
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany; Institute of Radiation Medicine (IRM), Department of Radiation Sciences, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Christian Schneeweis
- Department of Medicine II, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany
| | - Roland M Schmid
- Department of Medicine II, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany
| | - Günter Schneider
- Department of Medicine II, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany; Department of General Visceral and Pediatric Surgery, University Medical Center Göttingen, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany; Institute of Radiation Medicine (IRM), Department of Radiation Sciences, Helmholtz Zentrum Munich, Neuherberg, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Sophie Dobiasch
- Department of Radiation Oncology, School of Medicine, Klinikum rechts der Isar, Technical University Munich (TUM), Munich, Germany; Institute of Radiation Medicine (IRM), Department of Radiation Sciences, Helmholtz Zentrum Munich, Neuherberg, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
| |
Collapse
|
46
|
Adamopoulos C, Cave DD, Papavassiliou AG. Inhibition of the RAF/MEK/ERK Signaling Cascade in Pancreatic Cancer: Recent Advances and Future Perspectives. Int J Mol Sci 2024; 25:1631. [PMID: 38338909 PMCID: PMC10855714 DOI: 10.3390/ijms25031631] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Pancreatic cancer represents a formidable challenge in oncology, primarily due to its aggressive nature and limited therapeutic options. The prognosis of patients with pancreatic ductal adenocarcinoma (PDAC), the main form of pancreatic cancer, remains disappointingly poor with a 5-year overall survival of only 5%. Almost 95% of PDAC patients harbor Kirsten rat sarcoma virus (KRAS) oncogenic mutations. KRAS activates downstream intracellular pathways, most notably the rapidly accelerated fibrosarcoma (RAF)/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling axis. Dysregulation of the RAF/MEK/ERK pathway is a crucial feature of pancreatic cancer and therefore its main components, RAF, MEK and ERK kinases, have been targeted pharmacologically, largely by small-molecule inhibitors. The recent advances in the development of inhibitors not only directly targeting the RAF/MEK/ERK pathway but also indirectly through inhibition of its regulators, such as Src homology-containing protein tyrosine phosphatase 2 (SHP2) and Son of sevenless homolog 1 (SOS1), provide new therapeutic opportunities. Moreover, the discovery of allele-specific small-molecule inhibitors against mutant KRAS variants has brought excitement for successful innovations in the battle against pancreatic cancer. Herein, we review the recent advances in targeted therapy and combinatorial strategies with focus on the current preclinical and clinical approaches, providing critical insight, underscoring the potential of these efforts and supporting their promise to improve the lives of patients with PDAC.
Collapse
Affiliation(s)
- Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Donatella Delle Cave
- Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’, CNR, 80131 Naples, Italy
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
47
|
Benitz S, Steep A, Nasser M, Preall J, Mahajan UM, McQuithey H, Loveless I, Davis ET, Wen HJ, Long DW, Metzler T, Zwernik S, Louw M, Rempinski D, Salas-Escabillas D, Brender S, Song L, Huang L, Zhang Z, Steele NG, Regel I, Bednar F, Crawford HC. ROR2 regulates cellular plasticity in pancreatic neoplasia and adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.13.571566. [PMID: 38168289 PMCID: PMC10760092 DOI: 10.1101/2023.12.13.571566] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Cellular plasticity is a hallmark of pancreatic ductal adenocarcinoma (PDAC) starting from the conversion of normal cells into precancerous lesions to the progression of carcinoma subtypes associated with aggressiveness and therapeutic response. We discovered that normal acinar cell differentiation, maintained by the transcription factor Pdx1, suppresses a broad gastric cell identity that is maintained in metaplasia, neoplasia, and the classical subtype of PDAC in mouse and human. We have identified the receptor tyrosine kinase Ror2 as marker of a gastric metaplasia (SPEM)-like identity in the pancreas. Ablation of Ror2 in a mouse model of pancreatic tumorigenesis promoted a switch to a gastric pit cell identity that largely persisted through progression to the classical subtype of PDAC. In both human and mouse pancreatic cancer, ROR2 activity continued to antagonize the gastric pit cell identity, strongly promoting an epithelial to mesenchymal transition, conferring resistance to KRAS inhibition, and vulnerability to AKT inhibition.
Collapse
Affiliation(s)
- Simone Benitz
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Alec Steep
- Center of Translational Data Science, University of Chicago, Chicago, Illinois, USA
| | - Malak Nasser
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Jonathan Preall
- Cold Spring Harbor Laboratory Cancer Center, Cold Spring Harbor, New York, USA
| | - Ujjwal M. Mahajan
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Holly McQuithey
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Ian Loveless
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Erick T. Davis
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Hui-Ju Wen
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Daniel W. Long
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Thomas Metzler
- Comparative Experimental Pathology (CEP), Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Samuel Zwernik
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Michaela Louw
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Donald Rempinski
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | | | - Sydney Brender
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Linghao Song
- Center of Translational Data Science, University of Chicago, Chicago, Illinois, USA
| | - Ling Huang
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Zhenyu Zhang
- Center of Translational Data Science, University of Chicago, Chicago, Illinois, USA
| | - Nina G. Steele
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
- Department of Pathology, Wayne State University, Detroit, Michigan, USA
- Department of Pharmacology and Toxicology, Michigan State University, Lansing, Michigan, USA
- Department of Oncology, Wayne State University, Detroit, Michigan, USA
| | - Ivonne Regel
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Filip Bednar
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Howard C. Crawford
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, USA
- Department of Pharmacology and Toxicology, Michigan State University, Lansing, Michigan, USA
- Department of Oncology, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
48
|
Tripathi P, Kumari R, Pathak R. Drugging the undruggable: Advances in targeting KRAS signaling in solid tumors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 385:1-39. [PMID: 38663957 DOI: 10.1016/bs.ircmb.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Cancer remains the leading cause of global mortality, prompting a paradigm shift in its treatment and outcomes with the advent of targeted therapies. Among the most prevalent mutations in RAS-driven cancers, Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations account for approximately 86% of cases worldwide, particularly in lung, pancreatic, and colon cancers, contributing to poor prognosis and reduced overall survival. Despite numerous efforts to understand the biology of KRAS mutants and their pivotal role in cancer development, the lack of well-defined drug-binding pockets has deemed KRAS an "undruggable" therapeutic target, presenting significant challenges for researchers and clinicians alike. Through significant biochemical and technological advances, the last decade has witnessed promising breakthroughs in targeted therapies for KRAS-mutated lung, colon, and pancreatic cancers, marking a critical turning point in the field. In this chapter, we provide an overview of the characteristics of KRAS mutations across various solid tumors, highlighting ongoing cutting-edge research on the immune microenvironment, the development of KRAS-driven mice models, and the recent progress in the exploration of specific KRAS mutant-targeted therapeutic approaches. By comprehensive understanding of the intricacies of KRAS signaling in solid tumors and the latest therapeutic developments, this chapter will shed light on the potential for novel therapeutic strategies to combat KRAS-driven tumors and improve patient outcomes.
Collapse
Affiliation(s)
- Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, United States
| | - Rajni Kumari
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, United States.
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, United States.
| |
Collapse
|
49
|
Najumudeen AK, Fey SK, Millett LM, Ford CA, Gilroy K, Gunduz N, Ridgway RA, Anderson E, Strathdee D, Clark W, Nixon C, Morton JP, Campbell AD, Sansom OJ. KRAS allelic imbalance drives tumour initiation yet suppresses metastasis in colorectal cancer in vivo. Nat Commun 2024; 15:100. [PMID: 38168062 PMCID: PMC10762264 DOI: 10.1038/s41467-023-44342-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 12/09/2023] [Indexed: 01/05/2024] Open
Abstract
Oncogenic KRAS mutations are well-described functionally and are known to drive tumorigenesis. Recent reports describe a significant prevalence of KRAS allelic imbalances or gene dosage changes in human cancers, including loss of the wild-type allele in KRAS mutant cancers. However, the role of wild-type KRAS in tumorigenesis and therapeutic response remains elusive. We report an in vivo murine model of colorectal cancer featuring deletion of wild-type Kras in the context of oncogenic Kras. Deletion of wild-type Kras exacerbates oncogenic KRAS signalling through MAPK and thus drives tumour initiation. Absence of wild-type Kras potentiates the oncogenic effect of KRASG12D, while incidentally inducing sensitivity to inhibition of MEK1/2. Importantly, loss of the wild-type allele in aggressive models of KRASG12D-driven CRC significantly alters tumour progression, and suppresses metastasis through modulation of the immune microenvironment. This study highlights the critical role for wild-type Kras upon tumour initiation, progression and therapeutic response in Kras mutant CRC.
Collapse
Affiliation(s)
- Arafath K Najumudeen
- Cancer Research UK Scotland Institute, Glasgow, UK.
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Sigrid K Fey
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Laura M Millett
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Nuray Gunduz
- Cancer Research UK Scotland Institute, Glasgow, UK
| | | | - Eve Anderson
- Cancer Research UK Scotland Institute, Glasgow, UK
| | | | | | - Colin Nixon
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Jennifer P Morton
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Owen J Sansom
- Cancer Research UK Scotland Institute, Glasgow, UK.
- School of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
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
|