1
|
Anastasia A, Formenti L, Ostano P, Minoli L, Resovi A, Morosi L, Fioravanti C, Micotti E, Matteo C, Scanziani E, Chiorino G, Giavazzi R, Ghilardi C, Belotti D. Stroma gene signature predicts responsiveness to chemotherapy in pancreatic ductal adenocarcinoma patient-derived xenograft models. Mol Oncol 2025; 19:1075-1091. [PMID: 39902502 PMCID: PMC11977644 DOI: 10.1002/1878-0261.13816] [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: 07/23/2024] [Revised: 12/23/2024] [Accepted: 01/22/2025] [Indexed: 02/05/2025] Open
Abstract
Despite many efforts to understand the molecular mechanisms of pancreatic ductal adenocarcinoma (PDAC) treatment resistance, there is still no reliable method for selecting patients who could benefit from standard pharmacological treatment. Here, four PDAC patient-derived xenografts (PDAC-PDXs) with different responses to gemcitabine plus nab-paclitaxel (nanoparticle albumin-bound paclitaxel) were studied to dissect the contribution of both tumor and host microenvironment to treatment response. PDAC-PDXs transplanted into the pancreas of immunodeficient mice retained the main genetic and histopathological characteristics of the original human tumors, including invasiveness and desmoplastic reaction. Response to chemotherapy was associated with a specific 294 stroma gene signature and was not due to the intrinsic responsiveness of tumor cells or differences in drug delivery. Human dataset analysis validated the expression of the 294 stroma gene signature in PDAC clinical samples, confirming PDAC-PDXs as a useful tool to study the biology of tumor-host interactions and to test drug efficacy. In summary, we identified a stroma gene signature that differentiates PDAC-PDXs that are responsive to gemcitabine plus Nab-paclitaxel treatment from those that are not, confirming the active role of the tumor microenvironment in the drug response.
Collapse
Affiliation(s)
- Alessia Anastasia
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSBergamo and MilanItaly
| | - Laura Formenti
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSBergamo and MilanItaly
| | - Paola Ostano
- Lab of Cancer GenomicsFondazione “Edo ed Elvo Tempia”BiellaItaly
| | - Lucia Minoli
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSBergamo and MilanItaly
- Department of Veterinary Medicine and Animal Sciences (DIVAS)University of Milan (Unimi)LodiItaly
| | - Andrea Resovi
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSBergamo and MilanItaly
| | - Lavinia Morosi
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSBergamo and MilanItaly
- Present address:
IRCCS Humanitas Research HospitalRozzanoItaly
| | - Claudia Fioravanti
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSBergamo and MilanItaly
| | - Edoardo Micotti
- Department of NeuroscienceIstituto di Ricerche Farmacologiche Mario Negri IRCCSMilanItaly
| | - Cristina Matteo
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSBergamo and MilanItaly
| | - Eugenio Scanziani
- Mouse and Animal Pathology Laboratory (MAPLab), Fondazione UnimiMilanItaly
| | | | - Raffaella Giavazzi
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSBergamo and MilanItaly
| | - Carmen Ghilardi
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSBergamo and MilanItaly
| | - Dorina Belotti
- Department of OncologyIstituto di Ricerche Farmacologiche Mario Negri IRCCSBergamo and MilanItaly
| |
Collapse
|
2
|
Lo Buglio G, Lo Cicero A, Campora S, Ghersi G. The Multifaced Role of Collagen in Cancer Development and Progression. Int J Mol Sci 2024; 25:13523. [PMID: 39769286 PMCID: PMC11678882 DOI: 10.3390/ijms252413523] [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: 11/20/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/05/2025] Open
Abstract
Collagen is a crucial protein in the extracellular matrix (ECM) essential for preserving tissue architecture and supporting crucial cellular functions like proliferation and differentiation. There are twenty-eight identified types of collagen, which are further divided into different subgroups. This protein plays a critical role in regulating tissue homeostasis. However, in solid tumors, the balance can be disrupted, due to an abundance of collagen in the tumor microenvironment, which significantly affects tumor growth, cell invasion, and metastasis. It is important to investigate the specific types of collagens in cancer ECM and their distinct roles in tumor progression to comprehend their unique contribution to tumor behavior. The diverse pathophysiological functions of different collagen types in cancers illustrate collagen's dual roles, offering potential therapeutic options and serving as prognostic markers.
Collapse
Affiliation(s)
- Gabriele Lo Buglio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Alessandra Lo Cicero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
| | - Simona Campora
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
- Abiel srl, 90128 Palermo, Italy
| |
Collapse
|
3
|
Rosas J, Campanale JP, Harwood JL, Li L, Bae R, Cheng S, Tsou JM, Kaiser KM, Engle DD, Montell DJ, Pitenis AA. Differential Effects of Confinement on the Dynamics of Normal and Tumor-Derived Pancreatic Ductal Organoids. ACS APPLIED BIO MATERIALS 2024; 7:8489-8502. [PMID: 39576883 PMCID: PMC11653396 DOI: 10.1021/acsabm.4c01301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a cancer of the epithelia comprising the ductal network of the pancreas. During disease progression, PDAC tumors recruit fibroblasts that promote fibrosis, increasing local tissue stiffness and subjecting epithelial cells to increased compressive forces. Previous in vitro studies have documented cytoskeletal and nuclear adaptation following compressive stresses in two-dimensional (2D) and three-dimensional (3D) environments. However, a comparison of the responses of normal and tumor-derived ductal epithelia to physiologically relevant confinement remains underexplored, especially in 3D organoids. Here we control confinement with an engineered 3D microenvironment composed of Matrigel mixed with a low yield stress granular microgel. Normal and tumor-derived murine pancreas organoids (normal and tumor) were cultured for 48 h within this composite 3D environment or in pure Matrigel to investigate the effects of confinement on morphogenesis and lumen expansion. In confinement, tumor organoids (mT) formed a lumen that expanded rapidly, whereas normal organoids (mN) expanded more slowly. Moreover, a majority of normal organoids in more-confined conditions exhibited an inverted apicobasal polarity compared to those in less-confined conditions. Tumor organoids exhibited a collective "pulsing" behavior that increased in confinement. These pulses generated forces sufficient to locally overcome the yield stress of the microgels in the direction of organoid expansion. Normal organoids more commonly exhibit unidirectional rotation. Our in vitro microgel confinement platform enabled the discovery of two distinct modes of collective force generation in organoids that may shed light on the mutual interactions between tumors and the microenvironment. These insights into in vitro dynamics may deepen our understanding of how the confinement of healthy cells within a fibrotic tumor niche disrupts tissue organization and function in vivo.
Collapse
Affiliation(s)
- Jonah
M. Rosas
- Department
of Biomolecular Science & Engineering Program, University of California, Santa
Barbara, California 93106, United States
| | - Joseph P. Campanale
- Department
of Molecular, Cellular, and Developmental Biology, University of California, Santa
Barbara, California 93106, United States
| | - Jacob L. Harwood
- Department
of Molecular, Cellular, and Developmental Biology, University of California, Santa
Barbara, California 93106, United States
| | - Lufei Li
- Department
of Statistics and Applied Probability, University
of California, Santa Barbara, California 93106, United States
| | - Rachel Bae
- Department
of Chemistry & Biochemistry, University
of California, Santa Barbara, California 93106, United States
- Materials
Research Laboratory, University of California, Santa Barbara, California 93106, United States
| | - Shujun Cheng
- Department
of Molecular, Cellular, and Developmental Biology, University of California, Santa
Barbara, California 93106, United States
| | - Julia M. Tsou
- Department
of Molecular, Cellular, and Developmental Biology, University of California, Santa
Barbara, California 93106, United States
| | - Kathi M. Kaiser
- Department
of Experimental Physics, Saarland University, 66123 Saarbrücken, Germany
- Materials
Research Laboratory, University of California, Santa Barbara, California 93106, United States
| | - Dannielle D. Engle
- Salk Institute
for Biological Studies, La Jolla, California 92037, United States
| | - Denise J. Montell
- Department
of Biomolecular Science & Engineering Program, University of California, Santa
Barbara, California 93106, United States
- Department
of Molecular, Cellular, and Developmental Biology, University of California, Santa
Barbara, California 93106, United States
| | - Angela A. Pitenis
- Materials
Department, University of California, Santa Barbara, California 93106, United States
- Materials
Research Laboratory, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
4
|
Martinez ML, Nan K, Bao Z, Bacchetti R, Yuan S, Tyler J, Guezennec XL, Bard FA, Rainero E. Novel kinase regulators of extracellular matrix internalisation identified by high-content screening modulate invasive carcinoma cell migration. PLoS Biol 2024; 22:e3002930. [PMID: 39666682 PMCID: PMC11637276 DOI: 10.1371/journal.pbio.3002930] [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: 05/02/2024] [Accepted: 11/06/2024] [Indexed: 12/14/2024] Open
Abstract
The interaction between cancer cells and the extracellular matrix (ECM) plays a pivotal role in tumour progression. While the extracellular degradation of ECM proteins has been well characterised, ECM endocytosis and its impact on cancer cell progression, migration, and metastasis is poorly understood. ECM internalisation is increased in invasive breast cancer cells, suggesting it may support invasiveness. However, current high-throughput approaches mainly focus on cells grown on plastic in 2D, making it difficult to apply these to the study of ECM dynamics. Here, we developed a high-content screening assay to study ECM uptake, based on the of use automated ECM coating for the generation of highly homogeneous ECM a pH-sensitive dye to image ECM trafficking in live cells. We identified that mitogen-activated protein kinase (MAPK) family members, MAP3K1 and MAPK11 (p38β), and the protein phosphatase 2 (PP2) subunit PPP2R1A were required for the internalisation of ECM-bound α2β1 integrin. Mechanistically, we show that down-regulation of the sodium/proton exchanger 1 (NHE1), an established macropinocytosis regulator and a target of p38, mediated ECM macropinocytosis. Moreover, disruption of α2 integrin, MAP3K1, MAPK11, PPP2R1A, and NHE1-mediated ECM internalisation significantly impaired cancer cell migration and invasion in 2D and 3D culture systems. Of note, integrin-bound ECM was targeted for lysosomal degradation, which was required for cell migration on cell-derived matrices. Finally, α2β1 integrin and MAP3K1 expression were significantly up-regulated in pancreatic tumours and correlated with poor prognosis in pancreatic cancer patients. Strikingly, MAP3K1, MAPK11, PPP2R1A, and α2 integrin expression were higher in chemotherapy-resistant tumours in breast cancer patients. Our results identified the α2β1 integrin/p38 signalling axis as a novel regulator of ECM endocytosis, which drives invasive migration and tumour progression, demonstrating that our high-content screening approach has the capability of identifying novel regulators of cancer cell invasion.
Collapse
Affiliation(s)
- Montserrat Llanses Martinez
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Keqian Nan
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Zhe Bao
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Rachele Bacchetti
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Shengnan Yuan
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Joe Tyler
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | | | - Frederic A. Bard
- Institute of Molecular and Cell Biology, Singapore, Singapore
- Centre de Recherche en Cancérologie de Marseille, CRCM, Marseille, France
| | - Elena Rainero
- School of Biosciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
| |
Collapse
|
5
|
Walker M, Morton JP. Hydrogel models of pancreatic adenocarcinoma to study cell mechanosensing. Biophys Rev 2024; 16:851-870. [PMID: 39830124 PMCID: PMC11735828 DOI: 10.1007/s12551-024-01265-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is the predominant form of pancreatic cancer and one of the leading causes of cancer-related death worldwide, with an extremely poor prognosis after diagnosis. High mortality from PDAC arises partly due to late diagnosis resulting from a lack of early-stage biomarkers and due to chemotherapeutic drug resistance, which arises from a highly fibrotic stromal response known as desmoplasia. Desmoplasia alters tissue mechanics, which triggers changes in cell mechanosensing and leads to dysregulated transcriptional activity and disease phenotypes. Hydrogels are effective in vitro models to mimic mechanical changes in tissue mechanics during PDAC progression and to study the influence of these changes on mechanosensitive cell responses. Despite the complex biophysical changes that occur within the PDAC microenvironment, carefully designed hydrogels can very closely recapitulate these properties during PDAC progression. Hydrogels are relatively inexpensive, highly reproducible and can be designed in a humanised manner to increase their relevance for human PDAC studies. In vivo models have some limitations, including species-species differences, high variability, expense and legal/ethical considerations, which make hydrogel models a promising alternative. Here, we comprehensively review recent advancements in hydrogel bioengineering for developing our fundamental understanding of mechanobiology in PDAC, which is critical for informing advanced therapeutics.
Collapse
Affiliation(s)
- M Walker
- Centre for the Cellular Microenvironment, Advanced Research Centre, 11 Chapel Lane, James Watt School of Engineering, University of Glasgow, Glasgow, G11 6EW UK
| | - JP Morton
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Rd, Glasgow, G61 1BD UK
- School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow, G61 1QH UK
| |
Collapse
|
6
|
Rauth S, Malafa M, Ponnusamy MP, Batra SK. Emerging Trends in Gastrointestinal Cancer Targeted Therapies: Harnessing Tumor Microenvironment, Immune Factors, and Metabolomics Insights. Gastroenterology 2024; 167:867-884. [PMID: 38759843 PMCID: PMC11793124 DOI: 10.1053/j.gastro.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024]
Abstract
Gastrointestinal (GI) cancers are the leading cause of new cancer cases and cancer-related deaths worldwide. The treatment strategies for patients with GI tumors have focused on oncogenic molecular profiles associated with tumor cells. Recent evidence has demonstrated that the tumor cell functions are modulated by its microenvironment, compromising fibroblasts, extracellular matrices, microbiome, immune cells, and the enteric nervous system. Along with the tumor microenvironment components, alterations in key metabolic pathways have emerged as a hallmark of tumor cells. From these perspectives, this review will highlight the functions of different cellular components of the GI tumor microenvironment and their implications for treatment. Furthermore, we discuss the major metabolic reprogramming in GI tumor cells and how understanding metabolic rewiring could lead to new therapeutic strategies. Finally, we briefly summarize the targeted agents currently being studied in GI cancers. Understanding the complex interplay between tumor cell-intrinsic and -extrinsic factors during tumor progression is critical for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Mokenge Malafa
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, Nebraska.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, Nebraska.
| |
Collapse
|
7
|
Waldron RT, Lugea A, Chang HH, Su HY, Quiros C, Lewis MS, Che M, Ramanujan VK, Rozengurt E, Eibl G, Pandol SJ. Upregulated Matrisomal Proteins and Extracellular Matrix Mechanosignaling Underlie Obesity-Associated Promotion of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:1593. [PMID: 38672675 PMCID: PMC11048773 DOI: 10.3390/cancers16081593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Diet-induced obesity (DIO) promotes pancreatic ductal adenocarcinoma (PDAC) in mice expressing KRasG12D in the pancreas (KC mice), but the precise mechanisms remain unclear. Here, we performed multiplex quantitative proteomic and phosphoproteomic analysis by liquid chromatography-tandem mass spectrometry and further bioinformatic and spatial analysis of pancreas tissues from control-fed versus DIO KC mice after 3, 6, and 9 months. Normal pancreatic parenchyma and associated proteins were steadily eliminated and the novel proteins, phosphoproteins, and signaling pathways associated with PDAC tumorigenesis increased until 6 months, when most males exhibited cancer, but females did not. Differentially expressed proteins and phosphoproteins induced by DIO revealed the crucial functional role of matrisomal proteins, which implies the roles of upstream regulation by TGFβ, extracellular matrix-receptor signaling to downstream PI3K-Akt-mTOR-, MAPK-, and Yap/Taz activation, and crucial effects in the tumor microenvironment such as metabolic alterations and signaling crosstalk between immune cells, cancer-associated fibroblasts (CAFs), and tumor cells. Staining tissues from KC mice localized the expression of several prognostic PDAC biomarkers and elucidated tumorigenic features, such as robust macrophage infiltration, acinar-ductal metaplasia, mucinous PanIN, distinct nonmucinous atypical flat lesions (AFLs) surrounded by smooth muscle actin-positive CAFs, invasive tumors with epithelial-mesenchymal transition arising close to AFLs, and expanding deserted areas by 9 months. We next used Nanostring GeoMX to characterize the early spatial distribution of specific immune cell subtypes in distinct normal, stromal, and PanIN areas. Taken together, these data richly contextualize DIO promotion of Kras-driven PDAC tumorigenesis and provide many novel insights into the signaling pathways and processes involved.
Collapse
Affiliation(s)
- Richard T. Waldron
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Aurelia Lugea
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hui-Hua Chang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Hsin-Yuan Su
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Crystal Quiros
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Michael S. Lewis
- Department of Medicine and Department of Pathology & Laboratory Medicine, VA Greater Los Angeles Health System, Cedars-Sinai Medical Center, Los Angeles, CA 90073, USA;
| | - Mingtian Che
- Biobank and Research Pathology Resource, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - V. Krishnan Ramanujan
- Biobank and Research Pathology Resource, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Stephen J. Pandol
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
8
|
Wang Y, Kong Y, Yang Q, Zhong C, Zhou D. Identification of fibronectin type III domain containing 3B as a potential prognostic and therapeutic target for pancreatic cancer: a preliminary analysis. Eur J Med Res 2024; 29:221. [PMID: 38581008 PMCID: PMC10996089 DOI: 10.1186/s40001-024-01823-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Fibronectin type III domain containing 3B (FNDC3B), a member of the fibronectin type III domain-containing protein family, has been indicated in various malignancies. However, the precise role of FNDC3B in the progression of pancreatic cancer (PC) still remains to be elucidated. METHODS In this study, we integrated data from the National Center for Biotechnology Information, the Cancer Genome Atlas, Genotype-Tissue Expression database, and Gene Expression Omnibus datasets to analyze FNDC3B expression and its association with various clinicopathological parameters. Subsequently, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, along with Gene Set Enrichment Analysis (GSEA), single sample Gene Set Enrichment Analysis (ssGSEA) and estimate analysis were recruited to delve into the biological function and immune infiltration based on FNDC3B expression. Additionally, the prognostic estimation was conducted using Cox analysis and Kaplan-Meier analysis. Subsequently, a nomogram was constructed according to the result of Cox analysis to enhance the prognostic ability of FNDC3B. Finally, the preliminary biological function of FNDC3B in PC cells was explored. RESULTS The study demonstrated a significantly higher expression of FNDC3B in tumor tissues compared to normal pancreatic tissues, and this expression was significantly associated with various clinicopathological parameters. GSEA revealed the involvement of FNDC3B in biological processes and signaling pathways related to integrin signaling pathway and cell adhesion. Additionally, ssGSEA analysis indicated a positive correlation between FNDC3B expression and infiltration of Th2 cells and neutrophils, while showing a negative correlation with plasmacytoid dendritic cells and Th17 cells infiltration. Kaplan-Meier analysis further supported that high FNDC3B expression in PC patients was linked to shorter overall survival, disease-specific survival, and progression-free interval. However, although univariate analysis demonstrated a significant correlation between FNDC3B expression and prognosis in PC patients, this association did not hold true in multivariate analysis. Finally, our findings highlight the crucial role of FNDC3B expression in regulating proliferation, migration, and invasion abilities of PC cells. CONCLUSION Despite limitations, the findings of this study underscored the potential of FNDC3B as a prognostic biomarker and its pivotal role in driving the progression of PC, particularly in orchestrating immune responses.
Collapse
Affiliation(s)
- Yizhi Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yang Kong
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Qifan Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Cheng Zhong
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Dongkai Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
9
|
Liu Y, Murazzi I, Fuller AM, Pan H, Irizarry-Negron VM, Devine A, Katti R, Skuli N, Ciotti GE, Pak K, Pack MA, Simon MC, Weber K, Cooper K, Eisinger-Mathason TK. Sarcoma Cells Secrete Hypoxia-Modified Collagen VI to Weaken the Lung Endothelial Barrier and Promote Metastasis. Cancer Res 2024; 84:977-993. [PMID: 38335278 PMCID: PMC10984776 DOI: 10.1158/0008-5472.can-23-0910] [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: 03/23/2023] [Revised: 12/21/2023] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Intratumoral hypoxia correlates with metastasis and poor survival in patients with sarcoma. Using an impedance sensing assay and a zebrafish intravital microinjection model, we demonstrated here that the hypoxia-inducible collagen-modifying enzyme lysyl hydroxylase PLOD2 and its substrate collagen type VI (COLVI) weaken the lung endothelial barrier and promote transendothelial migration. Mechanistically, hypoxia-induced PLOD2 in sarcoma cells modified COLVI, which was then secreted into the vasculature. Upon reaching the apical surface of lung endothelial cells, modified COLVI from tumor cells activated integrin β1 (ITGβ1). Furthermore, activated ITGβ1 colocalized with Kindlin2, initiating their interaction with F-actin and prompting its polymerization. Polymerized F-actin disrupted endothelial adherens junctions and induced barrier dysfunction. Consistently, modified and secreted COLVI was required for the late stages of lung metastasis in vivo. Analysis of patient gene expression and survival data from The Cancer Genome Atlas (TCGA) revealed an association between the expression of both PLOD2 and COLVI and patient survival. Furthermore, high levels of COLVI were detected in surgically resected sarcoma metastases from patient lungs and in the blood of tumor-bearing mice. Together, these data identify a mechanism of sarcoma lung metastasis, revealing opportunities for therapeutic intervention. SIGNIFICANCE Collagen type VI modified by hypoxia-induced PLOD2 is secreted by sarcoma cells and binds to integrin β1 on endothelial cells to induce barrier dysfunction, which promotes sarcoma vascular dissemination and metastasis.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ashley M. Fuller
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Hehai Pan
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Valerie M Irizarry-Negron
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Ann Devine
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Rohan Katti
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolas Skuli
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- Department of Cell and Developmental Biology
- University of Pennsylvania, Philadelphia, PA, USA
| | - Gabrielle E. Ciotti
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Koreana Pak
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Michael A. Pack
- Perelman School of Medicine
- Department of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - M. Celeste Simon
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- Department of Cell and Developmental Biology
- University of Pennsylvania, Philadelphia, PA, USA
| | - Kristy Weber
- Penn Sarcoma Program
- Perelman School of Medicine
- Department of Orthopedic Surgery
- University of Pennsylvania, Philadelphia, PA, USA
| | - Kumarasen Cooper
- Department of Pathology & Laboratory Medicine
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - T.S. Karin Eisinger-Mathason
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Mahmood M, Liu EM, Shergold AL, Tolla E, Tait-Mulder J, Huerta-Uribe A, Shokry E, Young AL, Lilla S, Kim M, Park T, Boscenco S, Manchon JL, Rodríguez-Antona C, Walters RC, Springett RJ, Blaza JN, Mitchell L, Blyth K, Zanivan S, Sumpton D, Roberts EW, Reznik E, Gammage PA. Mitochondrial DNA mutations drive aerobic glycolysis to enhance checkpoint blockade response in melanoma. NATURE CANCER 2024; 5:659-672. [PMID: 38286828 PMCID: PMC11056318 DOI: 10.1038/s43018-023-00721-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 12/20/2023] [Indexed: 01/31/2024]
Abstract
The mitochondrial genome (mtDNA) encodes essential machinery for oxidative phosphorylation and metabolic homeostasis. Tumor mtDNA is among the most somatically mutated regions of the cancer genome, but whether these mutations impact tumor biology is debated. We engineered truncating mutations of the mtDNA-encoded complex I gene, Mt-Nd5, into several murine models of melanoma. These mutations promoted a Warburg-like metabolic shift that reshaped tumor microenvironments in both mice and humans, consistently eliciting an anti-tumor immune response characterized by loss of resident neutrophils. Tumors bearing mtDNA mutations were sensitized to checkpoint blockade in a neutrophil-dependent manner, with induction of redox imbalance being sufficient to induce this effect in mtDNA wild-type tumors. Patient lesions bearing >50% mtDNA mutation heteroplasmy demonstrated a response rate to checkpoint blockade that was improved by ~2.5-fold over mtDNA wild-type cancer. These data nominate mtDNA mutations as functional regulators of cancer metabolism and tumor biology, with potential for therapeutic exploitation and treatment stratification.
Collapse
Affiliation(s)
| | - Eric Minwei Liu
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | - Engy Shokry
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Alex L Young
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Sergio Lilla
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Minsoo Kim
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tricia Park
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sonia Boscenco
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Javier L Manchon
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Crístina Rodríguez-Antona
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER, Madrid, Spain
| | - Rowan C Walters
- Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, The University of York, York, UK
| | - Roger J Springett
- Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, The University of York, York, UK
| | - James N Blaza
- Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, The University of York, York, UK
| | | | - Karen Blyth
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Sara Zanivan
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Edward W Roberts
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ed Reznik
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Payam A Gammage
- Cancer Research UK Scotland Institute, Glasgow, UK.
- School of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
11
|
Zhu L, Sun Z, Dai M, Wu H, Wang X, Xu J, Xue H, Jin Z, Nickel MD, Guo J, Sack I. Tomoelastography and Pancreatic Extracellular Volume Fraction Derived From MRI for Predicting Clinically Relevant Postoperative Pancreatic Fistula. J Magn Reson Imaging 2024; 59:1074-1082. [PMID: 37209387 DOI: 10.1002/jmri.28788] [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: 02/22/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Pancreatic stiffness and extracellular volume fraction (ECV) are potential imaging biomarkers for pancreatic fibrosis. Clinically relevant postoperative fistula (CR-POPF) is one of the most severe complications after pancreaticoduodenectomy. Which imaging biomarker performs better for predicting the risk of CR-POPF remains unknown. PURPOSE To evaluate the diagnostic performance of ECV and tomoelastography-derived pancreatic stiffness for predicting the risk of CR-POPF in patients undergoing pancreaticoduodenectomy. STUDY TYPE Prospective. POPULATION Eighty patients who underwent multiparametric pancreatic MRI before pancreaticoduodenectomy, among whom 16 developed CR-POPF and 64 did not. FIELD STRENGTH/SEQUENCE 3 T/tomoelastography and precontrast and postcontrast T1 mapping of the pancreas. ASSESSMENT Pancreatic stiffness was measured on the tomographic c-map, and pancreatic ECV was calculated from precontrast and postcontrast T1 maps. Pancreatic stiffness and ECV were compared with histological fibrosis grading (F0-F3). The optimal cutoff values for predicting CR-POPF were determined, and the correlation between CR-POPF and imaging parameters was evaluated. STATISTICAL TESTS The Spearman's rank correlation and multivariate linear regression analysis was conducted. The receiver operating characteristic curve analysis and logistic regression analysis was performed. A double-sided P < 0.05 indicated a statistically significant difference. RESULTS Pancreatic stiffness and ECV both showed a significantly positive correlation with histological pancreatic fibrosis (r = 0.73 and 0.56, respectively). Patients with advanced pancreatic fibrosis had significantly higher pancreatic stiffness and ECV compared to those with no/mild fibrosis. Pancreatic stiffness and ECV were also correlated with each other (r = 0.58). Lower pancreatic stiffness (<1.38 m/sec), lower ECV (<0.28), nondilated main pancreatic duct (<3 mm) and pathological diagnosis other than pancreatic ductal adenocarcinoma were associated with higher risk of CR-POPF at univariate analysis, and pancreatic stiffness was independently associated with CR-POPF at multivariate analysis (odds ratio: 18.59, 95% confidence interval: 4.45, 77.69). DATA CONCLUSION Pancreatic stiffness and ECV were associated with histological fibrosis grading, and pancreatic stiffness was an independent predictor for CR-POPF. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY STAGE: 5.
Collapse
Affiliation(s)
- Liang Zhu
- Department of Radiology, Peking Union Medical College Hospital, Beijing, China
| | - Zhaoyong Sun
- Department of Radiology, Peking Union Medical College Hospital, Beijing, China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Huanwen Wu
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Xuan Wang
- Department of Radiology, Peking Union Medical College Hospital, Beijing, China
| | - Jia Xu
- Department of Radiology, Peking Union Medical College Hospital, Beijing, China
| | - Huadan Xue
- Department of Radiology, Peking Union Medical College Hospital, Beijing, China
| | - Zhengyu Jin
- Department of Radiology, Peking Union Medical College Hospital, Beijing, China
| | | | - Jing Guo
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ingolf Sack
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
12
|
Obaid G, Eroy M, Zhao J, Bano S, Mino-Kenudson M, Hasan T. Immunofluorescence profiling of collagen subtypes is a predictor of treatment outcomes in pancreatic cancer. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 250:112811. [PMID: 38000171 PMCID: PMC10841621 DOI: 10.1016/j.jphotobiol.2023.112811] [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: 09/08/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023]
Abstract
Desmoplasia in pancreatic ductal adenocarcinoma (PDAC) is characterized by elevated levels of tumor collagen. Desmoplasia restricts drug delivery in PDAC, contributes to treatment resistance, and is associated with poor survival outcomes. We have previously shown that photodynamic therapy (PDT)-based treatment remediates desmoplasia in orthotopic PDAC tumors by reducing second harmonic generation signals from collagen by >90% and by reducing collagen alignment by >103-fold [19]. Remediating desmoplasia correlated with improved survival outcomes in mice. To understand this phenomenon at a fundamental level, it is important to dissect the impact of therapy on collagen subtypes. In this study, we demonstrate that immunofluorescence profiling of collagen subtypes I, II, III and IV in PDAC tumors 72 h following multiple treatment regimens is predictive of long-term outcomes. Treatment regimens include nanoliposomal irinotecan chemotherapy (nal-IRI; akin to ONIVYDE™), a combination of nal-IRI chemotherapy with PDT encapsulated in a single photoactivable multi-inhibitor liposome (PMIL) and an EGFR-targeted PMIL construct (TPMIL). Results show that the relative tumor content of collagen I, II and III was inversely correlated with overall survival (P ≤ 0.0013, P ≤ 0.0001, P ≤ 0.0011, respectively), while, surprisingly, the relative tumor content of collagen IV was directly correlated with overall survival (P ≤ 0.0001). Similar relationships were observed between the relative tumor content of collagen subtypes and the residual tumor volume at day 88 following treatment. Considering that the relationship between collagen subtypes and treatment outcomes is observed across multiple treatment regimens, immunofluorescence profiling at 72 h following treatment appears to be predictive of tumor growth inhibition and survival in PDAC. Early immunofluorescence collagen subtype profiling may therefore aid in treatment personalization and may inform the dosimetry and scheduling of combination regimens for PDAC, such as chemotherapy and emerging PDT-based combinations, to maximize patient survival benefit.
Collapse
Affiliation(s)
- Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Menitte Eroy
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jie Zhao
- Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Shazia Bano
- Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Mari Mino-Kenudson
- Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Tayyaba Hasan
- Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
13
|
Gautam SK, Batra SK, Jain M. Molecular and metabolic regulation of immunosuppression in metastatic pancreatic ductal adenocarcinoma. Mol Cancer 2023; 22:118. [PMID: 37488598 PMCID: PMC10367391 DOI: 10.1186/s12943-023-01813-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/23/2023] [Indexed: 07/26/2023] Open
Abstract
Immunosuppression is a hallmark of pancreatic ductal adenocarcinoma (PDAC), contributing to early metastasis and poor patient survival. Compared to the localized tumors, current standard-of-care therapies have failed to improve the survival of patients with metastatic PDAC, that necessecitates exploration of novel therapeutic approaches. While immunotherapies such as immune checkpoint blockade (ICB) and therapeutic vaccines have emerged as promising treatment modalities in certain cancers, limited responses have been achieved in PDAC. Therefore, specific mechanisms regulating the poor response to immunotherapy must be explored. The immunosuppressive microenvironment driven by oncogenic mutations, tumor secretome, non-coding RNAs, and tumor microbiome persists throughout PDAC progression, allowing neoplastic cells to grow locally and metastasize distantly. The metastatic cells escaping the host immune surveillance are unique in molecular, immunological, and metabolic characteristics. Following chemokine and exosomal guidance, these cells metastasize to the organ-specific pre-metastatic niches (PMNs) constituted by local resident cells, stromal fibroblasts, and suppressive immune cells, such as the metastasis-associated macrophages, neutrophils, and myeloid-derived suppressor cells. The metastatic immune microenvironment differs from primary tumors in stromal and immune cell composition, functionality, and metabolism. Thus far, multiple molecular and metabolic pathways, distinct from primary tumors, have been identified that dampen immune effector functions, confounding the immunotherapy response in metastatic PDAC. This review describes major immunoregulatory pathways that contribute to the metastatic progression and limit immunotherapy outcomes in PDAC. Overall, we highlight the therapeutic vulnerabilities attributable to immunosuppressive factors and discuss whether targeting these molecular and immunological "hot spots" could improve the outcomes of PDAC immunotherapies.
Collapse
Affiliation(s)
- Shailendra K Gautam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
14
|
Mahmood M, Liu EM, Shergold AL, Tolla E, Tait-Mulder J, Huerta Uribe A, Shokry E, Young AL, Lilla S, Kim M, Park T, Manchon J, Rodríguez-Antona C, Walters RC, Springett RJ, Blaza JN, Zanivan S, Sumpton D, Roberts EW, Reznik E, Gammage PA. Tumour mitochondrial DNA mutations drive aerobic glycolysis to enhance checkpoint blockade. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.21.533091. [PMID: 36993533 PMCID: PMC10055208 DOI: 10.1101/2023.03.21.533091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The mitochondrial genome encodes essential machinery for respiration and metabolic homeostasis but is paradoxically among the most common targets of somatic mutation in the cancer genome, with truncating mutations in respiratory complex I genes being most over-represented1. While mitochondrial DNA (mtDNA) mutations have been associated with both improved and worsened prognoses in several tumour lineages1-3, whether these mutations are drivers or exert any functional effect on tumour biology remains controversial. Here we discovered that complex I-encoding mtDNA mutations are sufficient to remodel the tumour immune landscape and therapeutic resistance to immune checkpoint blockade. Using mtDNA base editing technology4 we engineered recurrent truncating mutations in the mtDNA-encoded complex I gene, Mt-Nd5, into murine models of melanoma. Mechanistically, these mutations promoted utilisation of pyruvate as a terminal electron acceptor and increased glycolytic flux without major effects on oxygen consumption, driven by an over-reduced NAD pool and NADH shuttling between GAPDH and MDH1, mediating a Warburg-like metabolic shift. In turn, without modifying tumour growth, this altered cancer cell-intrinsic metabolism reshaped the tumour microenvironment in both mice and humans, promoting an anti-tumour immune response characterised by loss of resident neutrophils. This subsequently sensitised tumours bearing high mtDNA mutant heteroplasmy to immune checkpoint blockade, with phenocopy of key metabolic changes being sufficient to mediate this effect. Strikingly, patient lesions bearing >50% mtDNA mutation heteroplasmy also demonstrated a >2.5-fold improved response rate to checkpoint inhibitor blockade. Taken together these data nominate mtDNA mutations as functional regulators of cancer metabolism and tumour biology, with potential for therapeutic exploitation and treatment stratification.
Collapse
Affiliation(s)
| | - Eric Minwei Liu
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | - Engy Shokry
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | - Sergio Lilla
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Minsoo Kim
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tricia Park
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - J.L. Manchon
- Centro Nacional de Investigaciones Oncológicas(CNIO), Madrid, Spain
| | - Crístina Rodríguez-Antona
- Centro Nacional de Investigaciones Oncológicas(CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER, Madrid, Spain
| | - Rowan C. Walters
- Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, The University of York, York, UK
| | - Roger J. Springett
- Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, The University of York, York, UK
| | - James N. Blaza
- Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, The University of York, York, UK
| | - Sara Zanivan
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, UK
| | | | - Edward W. Roberts
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, UK
| | - Ed Reznik
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Payam A. Gammage
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, UK
| |
Collapse
|
15
|
First person – Vasileios Papalazarou. J Cell Sci 2022. [DOI: 10.1242/jcs.260866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
ABSTRACT
First Person is a series of interviews with the first authors of a selection of papers published in Journal of Cell Science, helping researchers promote themselves alongside their papers. Vasileios Papalazarou is joint first author on ‘ Collagen VI expression is negatively mechanosensitive in pancreatic cancer cells and supports the metastatic niche’, published in JCS. Vasileios conducted the research described in this article while a PhD student in Laura Machesky's lab at the Cancer Research UK Beatson Institute, Glasgow, UK. He is now a postdoctoral fellow in the lab of Karen Vousden at the Francis Crick Institute, London, UK, investigating cancer metabolism, cell migration and metastasis.
Collapse
|