1
|
Lee J, McClure S, Weichselbaum RR, Mimee M. Designing live bacterial therapeutics for cancer. Adv Drug Deliv Rev 2025:115579. [PMID: 40228606 DOI: 10.1016/j.addr.2025.115579] [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: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Humans are home to a diverse community of bacteria, many of which form symbiotic relationships with their host. Notably, tumors can also harbor their own unique bacterial populations that can influence tumor growth and progression. These bacteria, which selectively colonize hypoxic and acidic tumor microenvironments, present a novel therapeutic strategy to combat cancer. Advancements in synthetic biology enable us to safely and efficiently program therapeutic drugs production in bacteria, further enhancing their potential. This review provides a comprehensive guide to utilizing bacteria for cancer treatment. We discuss key considerations for selecting bacterial strains, emphasizing their colonization efficiency, the delicate balance between safety and anti-tumor efficacy, and the availability of tools for genetic engineering. We also delve into strategies for precise spatiotemporal control of drug delivery to minimize adverse effects and maximize therapeutic impact, exploring recent examples of engineered bacteria designed to combat tumors. Finally, we address the underlying challenges and future prospects of bacterial cancer therapy. This review underscores the versatility of bacterial therapies and outlines strategies to fully harness their potential in the fight against cancer.
Collapse
Affiliation(s)
- Jaehyun Lee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Sandra McClure
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago 60637, USA; The Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| | - Mark Mimee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
2
|
Majo S, Redoute-Timonnier C, Lacour A, Challeat L, Epinette E, Teillon J, Grosset CF, Auguste P. Optimized zymogram protocol from 3D spheroid cultures to study MMP-2 and -9 activities in tumor cells. BMC Biotechnol 2025; 25:28. [PMID: 40217226 PMCID: PMC11987344 DOI: 10.1186/s12896-025-00961-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Three-dimensional spheroids are more representative of tumors than cell-cultured monolayers. As in tumors, gradients of oxygen, nutrients and wastes are found in spheroid cultures but not in classical cultured monolayers. On the other hand, cell-based assays on the latter are hardly applicable to spheroid cultures. Such is the case for zymogram assays, which are classically used to measure MMP-2 and MMP-9 activities, and for immunoblots to measure the phosphorylation of proteins involved in ligand-induced intracellular signaling in normal and tumor cells. In this study we used two renal cancer cell lines as models, the first derived from a pediatric rhabdoid tumor and the second from an adult clear cell renal cell carcinoma. Using these two cell lines, we successfully developed a simple inexpensive assay to measure MMP-2 and MMP-9 activities in spheroids established in the presence of methylcellulose. After washing, 1 to 5 spheroids were pooled and stimulated with collagen I for 24 h before analysis. MMP-2 and MMP-9 activities were measured in supernatants using a standard but enhanced zymogram assay. Both pro-MMP-9 and MMP-2 activities were detected in spheroids established from both cell lines. In contrast with our previous data using classical cultures monolayers, collagen I stimulation decreased pro-MMP-9 activity without affecting MMP-2 activity. On the other hand, we could not accurately measure AKT intracellular signaling pathways from spheroids stimulated with collagen I. Finally, we adapted our 3D protocol to analyze the MAPK/ERK pathway in kidney tumor cells following induction by EGF. In conclusion, this zymogram assay for analyzing MMP-2 and MMP-9 activities in spheroids paves the way for novel experimentations in tumor biology.
Collapse
Affiliation(s)
- Sandra Majo
- Univ. Bordeaux, INSERM, BRIC, U1312, MIRCADE team, Bordeaux, F-33000, France
| | | | - Aurelie Lacour
- Univ. Bordeaux, INSERM, BRIC, U1312, MIRCADE team, Bordeaux, F-33000, France
| | - Laurine Challeat
- Univ. Bordeaux, INSERM, BRIC, U1312, MIRCADE team, Bordeaux, F-33000, France
| | - Eva Epinette
- Univ. Bordeaux, INSERM, BRIC, U1312, MIRCADE team, Bordeaux, F-33000, France
| | - Jeremie Teillon
- Univ. Bordeaux, CNRS, INSERM, BIC, US4, UAR 3420, Bordeaux, F-33000, France
| | | | - Patrick Auguste
- Univ. Bordeaux, INSERM, BRIC, U1312, MIRCADE team, Bordeaux, F-33000, France.
| |
Collapse
|
3
|
Zhang Z, Cheng W, Li H, Li X, Zhang X, Li Y, Wang B. Lactate-depleted pillar[5]arene-based chiral supramolecular nanovesicles for L-glucose-mediated tumor-specific chemodynamic- and photodynamic-synergistic therapy. J Mater Chem B 2025. [PMID: 40208309 DOI: 10.1039/d5tb00436e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
The distinct interactions of D/L-glucose with cells and biological systems have garnered significant attention. However, the impact of chiral glucose-modified nanomaterials on cancer diagnosis and treatment remains largely unexplored. Here, based on the host-guest interaction between D-/L-glucose-modified pillar[5]arene (D-/L-CP5) serving as the host molecule and Fe-porphyrin derivatives (FeTPPNHC) acting as the guest, an acid-responsive chiral supramolecular vesicle was constructed for transporting lactate oxidases (LOx) (denoted as LOx@D-/L-CP5⊃FeTPPNHC), aiming to enhance chirality-mediated tumor-specific cascade chemodynamic therapy (CDT) and photodynamic therapy (PDT) through the depletion of lactic acid (LA). Surprisingly, the L-glucose-mediated chiral vesicles exhibit remarkable chirality recognition and lactate depletion capabilities, which were higher than the D-glucose-mediated chiral vesicles. Once internalized by cancer cells, L-supramolecular nanomicelles can directly consume LA to generate a considerable amount of H2O2, which can then be converted into ˙OH and 1O2. In vitro and in vivo studies demonstrate the high tumor specificity and therapeutic efficacy of LOx@LCP5⊃FeTPPNHC. The findings suggest that chiral glucose-modified nanomaterials hold great potential in targeted cancer treatment, paving the way for the development of innovative cancer therapeutics based on their unique interactions with biological systems.
Collapse
Affiliation(s)
- Zefan Zhang
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Wenyuan Cheng
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Hui Li
- Department of Cardiovascular Medicine, Affiliated Hospital of Yan'an University, Yan'an, 716000, P. R. China
| | - Xinxin Li
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Xuan Zhang
- Department of Cardiovascular Medicine, Affiliated Hospital of Yan'an University, Yan'an, 716000, P. R. China
| | - Yu Li
- Dr. Y. Li, Department of Magnetic Resonance, The First Clinical Medical College, Lanzhou University, Lanzhou, 730030, P. R. China
| | - Baodui Wang
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, China.
| |
Collapse
|
4
|
Ibe A, Nakanishi M, Higashimoto K, Muragaki Y, Ehata S. An acidic microenvironment promotes lymphatic metastasis of melanoma by Thy-1 in endothelial cells and integrin αvβ3 in tumor cells. Discov Oncol 2025; 16:498. [PMID: 40205271 PMCID: PMC11981969 DOI: 10.1007/s12672-025-02276-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Melanoma tissues exhibit an acidic microenvironment compared with that of surrounding normal tissues. However, the effects of acidic conditions on the lymphatic metastasis, a crucial prognostic factor for patients with melanoma, are unclear. In the present study, we aimed to investigate the role of the acidic microenvironment in the function of lymphatic endothelial cells. We first conducted gene expression profiling using human dermal lymphatic endothelial cells (HDLECs) treated with low pH media. Based on these results, we focused on Thy-1/CD90, whose expression increased in a time-dependent manner in HDLECs under acidic conditions. Immunohistochemical analysis of primary tumor tissues in a mouse melanoma model revealed an increased expression of Thy-1 in lymphatic endothelial cells. The expression of integrin αvβ3, a receptor for Thy-1, was also up-regulated in melanoma cells under acidic conditions. The adhesion of HDLECs to melanoma cells was accelerated under acidic conditions, which was reduced by Thy-1 knockdown in HDLECs. Furthermore, lymphatic metastasis was significantly attenuated in a mouse melanoma metastasis model when inoculated with integrin αv-silenced melanoma cells. These results suggest that acid-induced Thy-1 in lymphatic endothelial cells, as well as integrin αvβ3 in melanoma cells, may promote their mutual cellular adhesion, contributing to lymphatic metastasis.
Collapse
Affiliation(s)
- Akiya Ibe
- Department of Pathology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Masako Nakanishi
- Department of Pathology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| | - Kurumi Higashimoto
- Department of Pathology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Yasuteru Muragaki
- Department of Pathology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Shogo Ehata
- Department of Pathology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| |
Collapse
|
5
|
Gao F, Shah R, Xin G, Wang R. Metabolic Dialogue Shapes Immune Response in the Tumor Microenvironment. Eur J Immunol 2025; 55:e202451102. [PMID: 40223597 PMCID: PMC11995254 DOI: 10.1002/eji.202451102] [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: 10/24/2024] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/15/2025]
Abstract
The fate of immune cells is fundamentally linked to their metabolic program, which is also influenced by the metabolic landscape of their environment. The tumor microenvironment represents a unique system for intercellular metabolic interactions, where tumor-derived metabolites suppress effector CD8+ T cells and promote tumor-promoting macrophages, reinforcing an immune-suppressive niche. This review will discuss recent advancements in metabolism research, exploring the interplay between various metabolites and their effects on immune cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Fengxia Gao
- Department of Microbial Infection and ImmunityPelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOhioUSA
| | - Rushil Shah
- Center for Childhood Cancer ResearchHematology/Oncology & BMTAbigail Wexner Research Institute at Nationwide Children's HospitalDepartment of PediatricsThe Ohio State UniversityColumbusOhioUSA
| | - Gang Xin
- Department of Microbial Infection and ImmunityPelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOhioUSA
| | - Ruoning Wang
- Center for Childhood Cancer ResearchHematology/Oncology & BMTAbigail Wexner Research Institute at Nationwide Children's HospitalDepartment of PediatricsThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
6
|
Zhu Y, Zhou Z, Du X, Lin X, Liang ZM, Chen S, Sun Y, Wang Y, Na Z, Wu Z, Zhong J, Han B, Zhu X, Fu W, Li H, Luo ML, Hu H. Cancer cell-derived arginine fuels polyamine biosynthesis in tumor-associated macrophages to promote immune evasion. Cancer Cell 2025:S1535-6108(25)00116-3. [PMID: 40185095 DOI: 10.1016/j.ccell.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 01/08/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Arginine metabolism reshapes the tumor microenvironment (TME) into a pro-tumor niche through complex metabolic cross-feeding among various cell types. However, the key intercellular metabolic communication that mediates the collective effects of arginine metabolism within the TME remains unclear. Here, we reveal that the metabolic interplay between cancer cells and macrophages plays a dominant role in arginine-driven breast cancer progression. Within the TME, breast cancer cells serve as the primary source of arginine, which induces a pro-tumor polarization of tumor-associated macrophages (TAMs), thereby suppressing the anti-tumor activity of CD8+ T cells. Notably, this cancer cell-macrophage interaction overrides the arginine-mediated enhancement of CD8+ T cell anti-tumor activity. Mechanistically, polyamines derived from arginine metabolism enhance pro-tumor TAM polarization via thymine DNA glycosylase (TDG)-mediated DNA demethylation, regulated by p53 signaling. Importantly, targeting the arginine-polyamine-TDG axis between cancer cells and macrophages significantly suppresses breast cancer growth, highlighting its therapeutic potential.
Collapse
Affiliation(s)
- Yinghua Zhu
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Genetic Medicine, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Ziwei Zhou
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xin Du
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xiaorong Lin
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou 515031, China
| | - Zhi-Mei Liang
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Si Chen
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yiwei Sun
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310018, China; Experimental Research Center, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yue Wang
- Experimental Research Center, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Zhenkun Na
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310018, China
| | - Zhiyong Wu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou 515031, China
| | - Jiaxin Zhong
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Beinan Han
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xiangping Zhu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Wenkui Fu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Hongde Li
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310018, China.
| | - Man-Li Luo
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Hai Hu
- Breast Cancer Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310022, China.
| |
Collapse
|
7
|
Altea-Manzano P, Decker-Farrell A, Janowitz T, Erez A. Metabolic interplays between the tumour and the host shape the tumour macroenvironment. Nat Rev Cancer 2025; 25:274-292. [PMID: 39833533 DOI: 10.1038/s41568-024-00786-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
Metabolic reprogramming of cancer cells and the tumour microenvironment are pivotal characteristics of cancers, and studying these processes offer insights and avenues for cancer diagnostics and therapeutics. Recent advancements have underscored the impact of host systemic features, termed macroenvironment, on facilitating cancer progression. During tumorigenesis, these inherent features of the host, such as germline genetics, immune profile and the metabolic status, influence how the body responds to cancer. In parallel, as cancer grows, it induces systemic effects beyond the primary tumour site and affects the macroenvironment, for example, through inflammation, the metabolic end-stage syndrome of cachexia, and metabolic dysregulation. Therefore, understanding the intricate metabolic interplay between the tumour and the host is a growing frontier in advancing cancer diagnosis and therapy. In this Review, we explore the specific contribution of the metabolic fitness of the host to cancer initiation, progression and response to therapy. We then delineate the complex metabolic crosstalk between the tumour, the microenvironment and the host, which promotes disease progression to metastasis and cachexia. The metabolic relationships among the host, cancer pathogenesis and the consequent responsive systemic manifestations during cancer progression provide new perspectives for mechanistic cancer therapy and improved management of patients with cancer.
Collapse
Affiliation(s)
| | | | | | - Ayelet Erez
- Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
8
|
Gore M, Kabekkodu SP, Chakrabarty S. Exploring the metabolic alterations in cervical cancer induced by HPV oncoproteins: From mechanisms to therapeutic targets. Biochim Biophys Acta Rev Cancer 2025; 1880:189292. [PMID: 40037419 DOI: 10.1016/j.bbcan.2025.189292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
The role of human Papillomavirus (HPV) in metabolic reprogramming is implicated in the development and progression of cervical cancer. During carcinogenesis, cancer cells modify various metabolic pathways to generate energy and sustain their growth and development. Cervical cancer, one of the most prevalent malignancies affecting women globally, involves metabolic alterations such as increased glycolysis, elevated lactate production, and lipid accumulation. The oncoproteins, primarily E6 and E7, which are encoded by high-risk HPVs, facilitate the accumulation of several cancer markers, promoting not only the growth and development of cancer but also metastasis, immune evasion, and therapy resistance. HPV oncoproteins interact with cellular MYC (c-MYC), retinoblastoma protein (pRB), p53, and hypoxia-inducible factor 1α (HIF-1α), leading to the induction of metabolic reprogramming and favour the Warburg effect. Metabolic reprogramming enables HPV to persist for an extended period and accelerates the progression of cervical cancer. This review summarizes the role of HPV oncoproteins in metabolic reprogramming and their contributions to the development and progression of cervical cancer. Additionally, this review provides insights into how metabolic reprogramming opens avenues for novel therapeutic strategies, including the discovery of new and repurposed drugs that could be applied to treat cervical cancer.
Collapse
Affiliation(s)
- Mrudula Gore
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
9
|
Sencha LM, Karpova MA, Dobrynina OE, Balalaeva IV. Cell-type dependent effect of 3D collagen matrix on cancer cell resistance to suboptimal conditions: the case of serum deprivation, glucose starvation, and hypoxia. Tissue Cell 2025; 93:102719. [PMID: 39823703 DOI: 10.1016/j.tice.2024.102719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/13/2024] [Accepted: 12/29/2024] [Indexed: 01/20/2025]
Abstract
The extracellular matrix (ECM) and its primary chemical components, including collagen, play a pivotal role in carcinogenesis and tumor progression. The ECM actively regulates cell proliferation, migration, and, importantly, resistance to various adverse factors. It is widely recognized as a key factor in modifying the resistance of tumor cells to various treatment modalities and cytotoxic compounds. However, the role of the ECM in tumor cell adaptation to nutritional deficiencies and hypoxic conditions remains significantly less studied. Since it is generally accepted that tumor cells resistance increases when cultured in a three-dimensional matrix, we sought to experimentally test the universality of this statement. In this work, we analyzed the responses of tumor cells with varying origins and proliferative activities, including human bladder carcinoma, epidermoid carcinoma, and ovarian carcinoma, to deprivation of serum, glucose and oxygen. We compared cell resistance to suboptimal conditions when cultured in a monolayer on tissue culture (TC)-treated polystyrene, on collagen-coated surfaces, or within a three-dimensional hydrogel composed of collagen type I. All three cell lines were stably transfected with fluorescent protein genes. To register the cell growth dynamics, we used a fluorescence-based technique that allows long-term quantitative observations without disrupting the hydrogel. The analyzed cell lines demonstrated different patterns of relative sensitivity to suboptimal conditions. We revealed that the direction and intensity of the collagen matrix effect depend on the cell type. Slowly proliferating ovarian carcinoma cells showed no noticeable changes in their behavior when cultured in a gel compared to a monolayer. In the case of bladder carcinoma, we registered predominantly resistance-stimulating effect of the collagen matrix, but it was significant only under serum deprivation. The most pronounced effect of collagen was registered for epidermoid carcinoma. Importantly, this effect was ambivalent: gel-embedded cells demonstrated significantly enhanced resistance to serum deprivation, but, at the same time, they were more responsive to glucose starvation and hypoxic conditions. We attribute the registered phenomenon to the individual characteristics of tumor cells with different origins and metabolic activities.
Collapse
Affiliation(s)
- Ludmila M Sencha
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Maria A Karpova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Olga E Dobrynina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Irina V Balalaeva
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.
| |
Collapse
|
10
|
Drapela S, Garcia BM, Gomes AP, Correia AL. Metabolic landscape of disseminated cancer dormancy. Trends Cancer 2025; 11:321-333. [PMID: 39510896 PMCID: PMC11981868 DOI: 10.1016/j.trecan.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024]
Abstract
Cancer dormancy is a phenomenon defined by the entry of cancer cells into a reversible quiescent, nonproliferative state, and represents an essential part of the metastatic cascade responsible for cancer recurrence and mortality. Emerging evidence suggests that metabolic reprogramming plays a pivotal role in enabling entry, maintenance, and exit from dormancy in the face of the different environments of the metastatic cascade. Here, we review the current literature to understand the dynamics of metabolism during dormancy, highlighting its fine-tuning by the host micro- and macroenvironment, and put forward the importance of identifying metabolic vulnerabilities of the dormant state as therapeutic targets to eradicate recurrent disease.
Collapse
Affiliation(s)
- Stanislav Drapela
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Bruna M Garcia
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Ana P Gomes
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | | |
Collapse
|
11
|
Guo S, Zhang L, Ren J, Lu Z, Ma X, Liu X, Jin H, Li J. The roles of enhancer, especially super-enhancer-driven genes in tumor metabolism and immunity. Int J Biol Macromol 2025; 308:142414. [PMID: 40132720 DOI: 10.1016/j.ijbiomac.2025.142414] [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: 10/16/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Abnormal metabolism is a characteristic of malignant tumors. Numerous factors play roles in the regulation of tumor metabolism. As epigenetic regulators, enhancers, especially the super-enhancers (SEs), serve as platforms for transcription factors that regulate the expression of metabolism-related enzymes or transporters at the gene level. In this study, we review the effects of enhancer/ SE-driven genes on tumor metabolism and immunity. Enhancers/SEs play regulatory roles in glucose metabolism (glycolysis, gluconeogenesis, tricarboxylic acid (TCA) cycle, pyruvate, and pentose phosphate pathway, lipid metabolism (cholesterol, fatty acid, phosphatide, and sphingolipid), and amino acid metabolism (glutamine, tryptophan, arginine, and cystine). By regulating tumor metabolism, enhancers and SEs can reprogram tumor microenvironment, especially the status of various immune cells. Therefore, interfering enhancers/SEs that regulate the tumor metabolism is likely to enhance the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Songyue Guo
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Lu Zhang
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Jiao Ren
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Zhong Lu
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Xiaolin Ma
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Xinling Liu
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China.
| | - Hongchuan Jin
- Department of Medical Oncology, Cancer Center of Zhejiang University, Sir Run Run Shaw hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, China.
| | - Jiaqiu Li
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China.
| |
Collapse
|
12
|
Xiao J, Liu T, Zeng F, Zhang J. New insights into T cell metabolism in liver cancer: from mechanism to therapy. Cell Death Discov 2025; 11:118. [PMID: 40122853 PMCID: PMC11930970 DOI: 10.1038/s41420-025-02397-w] [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: 07/08/2024] [Revised: 02/19/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
Liver cancer is the sixth most common cancer worldwide and the third most common cause of cancer mortality. The development and progression of liver cancer and metastases is a multifaceted process involving numerous metabolic pathways. T cells have a protective role in the defense against cancer, and manipulating metabolic pathways in T cells can alter their antitumor activity. Furthermore, Liver cancer and T cell nutrition competition lead to T cell dysfunction through various molecular mechanisms. Some nanomaterials and drugs can improve T cell metabolism and promote the anti-liver cancer function of T cells. This review discusses the current literature regarding metabolic changes in liver cancer, the role of T cells in liver cancer, T cell metabolism in liver cancer, and targeted T cell metabolism therapy for liver cancer. The promise and challenges of studying target T cell metabolism for treating liver cancer are also addressed. Targeting T cell metabolism is a promising approach for treating liver cancer.
Collapse
Affiliation(s)
- Jie Xiao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China
| | - Ting Liu
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichua, China.
| | - Jinhua Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China.
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China.
| |
Collapse
|
13
|
Liu Y, Wan L, Chen Y, Zhang R, Xia Y, Chen M, Huang X, Liu R. Fatty acid metabolism related gene MECR contributes to the progression of prostate cancer. Cancer Cell Int 2025; 25:105. [PMID: 40108606 PMCID: PMC11924791 DOI: 10.1186/s12935-025-03738-6] [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: 03/28/2024] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the most common urological malignancy and second only to lung cancer in incidence among men. Its prognosis varies widely due to its heterogeneity. Research indicates that fatty acid metabolism may play a role in tumor development. METHODS The gene expression profiles of PCa cell lines (GSE6919) in GEO database were analyzed to identify differentially expressed genes and their significance in relation to progression-free interval. The R package was employed to assess overall survival significance and clinicopathological features. The study investigated the effects of gene mutations and methylation on PCa and their correlation with immune cell infiltration in the tumor microenvironment, utilizing cBioPortal and UALCAN resources. TIMER was used in the TCGA project to compare the expression of MECR in tumours and in adjacent normal tissue for different tumours or for specific tumour subtypes. Furthermore, we examined the impact of hub genes on PCa progression through RT qPCR, immunohistochemistry, and cellular assays. RESULTS The MECR gene, which plays a role in fatty acid metabolism, has been implicated in the development and progression of PCa. Its expression levels are significantly associated with clinical features, survival outcomes, and prognosis in PCa. Comprehensive analyses of MECR mutations and methylation levels further underscore its involvement in the progression of prostate cancer. Additionally, MECR is closely associated with the immune microenvironment and immune cell infiltration in PCa. Furthermore, the in vitro and in vivo data indicated that MECR plays a role in PCa proliferation, migration, and invasion. CONCLUSION MECR has significant potential for research and application in the assessment of PCa prognosis and the regulation of the immune microenvironment.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, China
| | - Lilin Wan
- Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, China
| | - Yuxuan Chen
- Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, China
| | - Ruixin Zhang
- Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, China
| | - Yi Xia
- Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, China
| | - Ming Chen
- Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, China
| | - Xiang Huang
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Ruiji Liu
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
14
|
Jonker PB, Sadullozoda M, Cognet G, Saab JJA, Sokol KH, Wu VX, Kumari D, Sheehan C, Ozgurses ME, Agovino D, Croley G, Patel SA, Bock-Hughes A, Macleod KF, Shah H, Coloff JL, Lien EC, Muir A. Microenvironmental arginine restriction sensitizes pancreatic cancers to polyunsaturated fatty acids by suppression of lipid synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642426. [PMID: 40161789 PMCID: PMC11952453 DOI: 10.1101/2025.03.10.642426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Nutrient limitation is a characteristic feature of poorly perfused tumors. In contrast to well-perfused tissues, nutrient deficits in tumors perturb cellular metabolic activity, which imposes metabolic constraints on cancer cells. The metabolic constraints created by the tumor microenvironment can lead to vulnerabilities in cancers. Identifying the metabolic constraints of the tumor microenvironment and the vulnerabilities that arise in cancers can provide new insight into tumor biology and identify promising antineoplastic targets. To identify how the microenvironment constrains the metabolism of pancreatic tumors, we challenged pancreatic cancer cells with microenvironmental nutrient levels and analyzed changes in cell metabolism. We found that arginine limitation in pancreatic tumors perturbs saturated and monounsaturated fatty acid synthesis by suppressing the lipogenic transcription factor SREBP1. Synthesis of these fatty acids is critical for maintaining a balance of saturated, monounsaturated, and polyunsaturated fatty acids in cellular membranes. As a consequence of microenvironmental constraints on fatty acid synthesis, pancreatic cancer cells and tumors are unable to maintain lipid homeostasis when exposed to polyunsaturated fatty acids, leading to cell death by ferroptosis. In sum, arginine restriction in the tumor microenvironment constrains lipid metabolism in pancreatic cancers, which renders these tumors vulnerable to polyunsaturated-enriched fat sources.
Collapse
Affiliation(s)
- Patrick B. Jonker
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Mumina Sadullozoda
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Guillaume Cognet
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Juan J. Apiz Saab
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Kelly H. Sokol
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, Michigan, USA, 49503
| | - Violet X. Wu
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Deepa Kumari
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Colin Sheehan
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Mete E. Ozgurses
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, USA, 60612
| | - Darby Agovino
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Grace Croley
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Smit A. Patel
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Althea Bock-Hughes
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Kay F. Macleod
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| | - Hardik Shah
- Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA, 60637
| | - Jonathan L. Coloff
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL, USA, 60612
| | - Evan C. Lien
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, Michigan, USA, 49503
| | - Alexander Muir
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA, 60637
| |
Collapse
|
15
|
Cabezón-Gutiérrez L, Palka-Kotlowska M, Custodio-Cabello S, Chacón-Ovejero B, Pacheco-Barcia V. Metabolic mechanisms of immunotherapy resistance. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002297. [PMID: 40092297 PMCID: PMC11907103 DOI: 10.37349/etat.2025.1002297] [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: 12/08/2024] [Accepted: 02/22/2025] [Indexed: 03/19/2025] Open
Abstract
Immunotherapy has revolutionized cancer treatment, yet its efficacy is frequently compromised by metabolic mechanisms that drive resistance. Understanding how tumor metabolism shapes the immune microenvironment is essential for developing effective therapeutic strategies. This review examines key metabolic pathways influencing immunotherapy resistance, including glucose, lipid, and amino acid metabolism. We discuss their impact on immune cell function and tumor progression, highlighting emerging therapeutic strategies to counteract these effects. Tumor cells undergo metabolic reprogramming to sustain proliferation, altering the availability of essential nutrients and generating toxic byproducts that impair cytotoxic T lymphocytes (CTLs) and natural killer (NK) cell activity. The accumulation of lactate, deregulated lipid metabolism, and amino acid depletion contribute to an immunosuppressive tumor microenvironment (TME). Targeting metabolic pathways, such as inhibiting glycolysis, modulating lipid metabolism, and restoring amino acid balance, has shown promise in enhancing immunotherapy response. Addressing metabolic barriers is crucial to overcoming immunotherapy resistance. Integrating metabolic-targeted therapies with immune checkpoint inhibitors may improve clinical outcomes. Future research should focus on personalized strategies to optimize metabolic interventions and enhance antitumor immunity.
Collapse
Affiliation(s)
- Luis Cabezón-Gutiérrez
- Medical Oncology, Hospital Universitario De Torrejón, 28850 Madrid, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Magda Palka-Kotlowska
- Medical Oncology, Hospital Universitario De Torrejón, 28850 Madrid, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Sara Custodio-Cabello
- Medical Oncology, Hospital Universitario De Torrejón, 28850 Madrid, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Beatriz Chacón-Ovejero
- Department of Pharmacy and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Vilma Pacheco-Barcia
- Medical Oncology, Hospital Universitario De Torrejón, 28850 Madrid, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| |
Collapse
|
16
|
Tang L, Peng S, Zhuang X, He Y, Song Y, Nie H, Zheng C, Pan Z, Lam AK, He M, Shi X, Li B, Xu WW. Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention. MEDCOMM – ONCOLOGY 2025; 4. [DOI: 10.1002/mog2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/10/2025] [Indexed: 03/04/2025]
Abstract
ABSTRACTMetastasis remains a leading cause of cancer‐related deaths, defined by a complex, multi‐step process in which tumor cells spread and form secondary growths in distant tissues. Despite substantial progress in understanding metastasis, the molecular mechanisms driving this process and the development of effective therapies remain incompletely understood. Elucidating the molecular pathways governing metastasis is essential for the discovery of innovative therapeutic targets. The rapid advancements in sequencing technologies and the expansion of biological databases have significantly deepened our understanding of the molecular drivers of metastasis and associated drug resistance. This review focuses on the molecular drivers of metastasis, particularly the roles of genetic mutations, epigenetic changes, and post‐translational modifications in metastasis progression. We also examine how the tumor microenvironment influences metastatic behavior and explore emerging therapeutic strategies, including targeted therapies and immunotherapies. Finally, we discuss future research directions, stressing the importance of novel treatment approaches and personalized strategies to overcome metastasis and improve patient outcomes. By integrating contemporary insights into the molecular basis of metastasis and therapeutic innovation, this review provides a comprehensive framework to guide future research and clinical advancements in metastatic cancer.
Collapse
Affiliation(s)
- Lin Tang
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Shao‐Cong Peng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Xiao‐Wan Zhuang
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Yan He
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Yu‐Xiang Song
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Hao Nie
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Can‐Can Zheng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Zhen‐Yu Pan
- Department of Radiation Oncology, The Affiliated Huizhou Hospital Guangzhou Medical University Huizhou China
| | - Alfred King‐Yin Lam
- Cancer Molecular Pathology and Griffith Medical School Griffith University Gold Coast Queensland Australia
| | - Ming‐Liang He
- Department of Biomedical Sciences City University of Hong Kong Hong Kong China
| | - Xing‐Yuan Shi
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Bin Li
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Wen Wen Xu
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| |
Collapse
|
17
|
Nelson AB, Reese LE, Rono E, Queathem ED, Qiu Y, McCluskey BM, Crampton A, Conniff E, Cummins K, Boytim E, Dansou S, Hwang J, Safo SE, Puchalska P, Wood DK, Schwertfeger KL, Crawford PA. Deciphering Colorectal Cancer-Hepatocyte Interactions: A Multiomics Platform for Interrogation of Metabolic Crosstalk in the Liver-Tumor Microenvironment. Int J Mol Sci 2025; 26:1976. [PMID: 40076609 PMCID: PMC11900982 DOI: 10.3390/ijms26051976] [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: 12/30/2024] [Revised: 02/01/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to adapt to and exploit their microenvironment for sustained growth. The liver is a common site of metastasis, but the interactions between tumor cells and hepatocytes remain poorly understood. In the context of liver metastasis, these interactions play a crucial role in promoting tumor survival and progression. This study leverages multiomics coverage of the microenvironment via liquid chromatography and high-resolution, high-mass-accuracy mass spectrometry-based untargeted metabolomics, 13C-stable isotope tracing, and RNA sequencing to uncover the metabolic impact of co-localized primary hepatocytes and a colon adenocarcinoma cell line, SW480, using a 2D co-culture model. Metabolic profiling revealed disrupted Warburg metabolism with an 80% decrease in glucose consumption and 94% decrease in lactate production by hepatocyte-SW480 co-cultures relative to SW480 control cultures. Decreased glucose consumption was coupled with alterations in glutamine and ketone body metabolism, suggesting a possible fuel switch upon co-culturing. Further, integrated multiomics analysis indicates that disruptions in metabolic pathways, including nucleoside biosynthesis, amino acids, and TCA cycle, correlate with altered SW480 transcriptional profiles and highlight the importance of redox homeostasis in tumor adaptation. Finally, these findings were replicated in three-dimensional microtissue organoids. Taken together, these studies support a bioinformatic approach to study metabolic crosstalk and discovery of potential therapeutic targets in preclinical models of the tumor microenvironment.
Collapse
Affiliation(s)
- Alisa B. Nelson
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA (E.D.Q.)
| | - Lyndsay E. Reese
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (L.E.R.)
| | - Elizabeth Rono
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Eric D. Queathem
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA (E.D.Q.)
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Yinjie Qiu
- Minnesota Supercomputing Institute, Minneapolis, MN 55455, USA
| | | | - Alexandra Crampton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Eric Conniff
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Katherine Cummins
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Ella Boytim
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Senali Dansou
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Justin Hwang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sandra E. Safo
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, MN 55455, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA (E.D.Q.)
| | - David K. Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Kathryn L. Schwertfeger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (L.E.R.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA (E.D.Q.)
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
18
|
Dsouza L, Pant A, Pope B, Yang Z. Vaccinia growth factor-dependent modulation of the mTORC1-CAD axis upon nutrient restriction. J Virol 2025; 99:e0211024. [PMID: 39817770 PMCID: PMC11852859 DOI: 10.1128/jvi.02110-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025] Open
Abstract
The molecular mechanisms by which vaccinia virus (VACV), the prototypical member of the poxviridae family, reprograms host cell metabolism remain largely unexplored. Additionally, cells sense and respond to fluctuating nutrient availability, thereby modulating metabolic pathways to ensure cellular homeostasis. Understanding how VACV modulates metabolic pathways in response to nutrient signals is crucial for understanding viral replication mechanisms, with the potential for developing antiviral therapies. In this study, we establish the importance of de novo pyrimidine synthesis during VACV infection. We report the significance of vaccinia growth factor (VGF), a viral early protein and a homolog of cellular epidermal growth factor (EGF), in enabling VACV to phosphorylate the key enzyme CAD of the de novo pyrimidine pathway at serine 1859, a site known to positively regulate CAD activity. Although nutrient-poor conditions typically inhibit mTORC1 activation, VACV activates CAD via the mTORC1-S6K1 signaling axis in a VGF-dependent manner, especially upon glutamine and asparagine limitation. However, unlike its cellular homolog EGF, the VGF peptide alone, in the absence of VACV infection, has minimal ability to activate CAD. This suggests the involvement of other viral factors yet to be identified. Our research provides a foundation for understanding the regulation of a significant metabolic pathway, de novo pyrimidine synthesis during VACV infection, shedding new light on viral regulation under distinct nutritional environments. This study not only has the potential to contribute to the advancement of antiviral treatments but also improve the development of VACV as an oncolytic agent and vaccine vector.IMPORTANCEViruses often reprogram host cell metabolism to facilitate replication. How poxviruses, such as the prototype member, vaccinia virus (VACV), modulate host cell metabolism is not well understood. Understanding how VACV affects these metabolic pathways is key to learning about viral replication and developing antiviral treatments. This study highlights the importance of de novo pyrimidine synthesis during VACV infection. We found that the vaccinia growth factor (VGF), a viral protein similar to the cellular epidermal growth factor (EGF), helps VACV activate the enzyme CAD of the de novo pyrimidine pathway. Upon nutrient limitation, VGF is needed for the activation of CAD through mTORC1-S6K signaling. VGF peptide alone is unable to activate this pathway independent of infection, suggesting the involvement of other viral factor(s). Our research not only sheds light on how VACV regulates metabolism but also holds promise for improving VACV as a cancer treatment and vaccine.
Collapse
Affiliation(s)
- Lara Dsouza
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Anil Pant
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Blake Pope
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Zhilong Yang
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
19
|
Xu Q, Hua X, Li B, Jiang B, Jin J, Wu R, Gu Y, Xu H, Cheng Q, Zhu S, Zhang F, Lv T, Song Y. Intrinsic STING of CD8 + T cells regulates self-metabolic reprogramming and memory to exert anti-tumor effects. Cell Commun Signal 2025; 23:99. [PMID: 39972350 PMCID: PMC11837649 DOI: 10.1186/s12964-025-02069-3] [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: 11/20/2024] [Accepted: 01/28/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Our team has previously found that the stimulator of interferon genes (STING) plays a more significant anti-tumor role in host immune cells than in tumor cells. Although STING is necessary for CD8 + T cells to exert immunological activity, its effect on CD8 + T cells remains debatable. In this study, we used both in vitro and in vivo models to explore the metabolic effects of STING on CD8 + T cells. METHODS Peripheral blood lymphocytes were procured from non-small cell lung cancer (NSCLC) patients receiving anti-PD-1 therapy to investigate the correlation between STING expression levels, CD8 + T-cell subsets, and immunotherapy efficacy. STING knockout (STING-KO) mice were used for in vivo studies. RNA-seq, seahorse, flow cytometry, electron microscopy, qPCR, immunofluorescence, western blotting, and immunoprecipitation were performed to explore the underlying mechanisms of STING in regulating CD8 + T cell function. RESULTS We discovered that the expression level of STING in immune cells exhibited a significant correlation with immunotherapy efficacy, as well as with the proportion of central memory CD8 + T cells. Moreover, we found that the loss of the STING gene results in a reduction in the number of mitochondria and a change in the metabolic pathway selection, thereby inducing excessive glycolysis in CD8 + T cells. This excessive glycolysis generates high levels of lactate, which further inhibits IFN-γ secretion and impacts memory T cell differentiation. Correcting the glycolysis disorder partially restored function and IFN-γ secretion, rescued the central memory CD8 + T subset, and improved immunotherapy in STING-KO mice. This provides a new treatment strategy for patients with low STING expression and a poor response to immunotherapy. CONCLUSION Intrinsic STING of CD8 + T cells affects their function through the HK2/Lactate/IFN-γ axis and affects memory differentiation by regulating glycolysis.
Collapse
Affiliation(s)
- Qiuli Xu
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210002, China
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Nanjing, Jiangsu, 210002, China
| | - Xin Hua
- Department of Geriatric Medicine, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Bingbing Li
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, School of Medicine, Nanjing University of Traditional Chinese Medicine Southeast University, #305 East Zhongshan Road, Nanjing, Jiangsu, 210002, China
| | - Bei Jiang
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, School of Medicine, Nanjing University of Traditional Chinese Medicine Southeast University, #305 East Zhongshan Road, Nanjing, Jiangsu, 210002, China
| | - Jiajia Jin
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Nanjing, Jiangsu, 210002, China
| | - Ranpu Wu
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210002, China
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Nanjing, Jiangsu, 210002, China
| | - Yanli Gu
- Department of Respiratory and Critical Care Medicine People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China
| | - Hao Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Nanjing, Jiangsu, 210002, China
- Nanjing Medical University, Nanjing, Jiangsu, 210002, China
| | - Qinpei Cheng
- Medical School of Nanjing University, Nanjing, Jiangsu, 210002, China
| | - Suhua Zhu
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Nanjing, Jiangsu, 210002, China
| | - Fang Zhang
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Nanjing, Jiangsu, 210002, China
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, School of Medicine, Nanjing University of Traditional Chinese Medicine Southeast University, #305 East Zhongshan Road, Nanjing, Jiangsu, 210002, China
| | - Tangfeng Lv
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, School of Medicine, Nanjing University of Traditional Chinese Medicine Southeast University, #305 East Zhongshan Road, Nanjing, Jiangsu, 210002, China.
| | - Yong Song
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210002, China.
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Nanjing, Jiangsu, 210002, China.
| |
Collapse
|
20
|
Chen J, Huang Z, Chen Y, Tian H, Chai P, Shen Y, Yao Y, Xu S, Ge S, Jia R. Lactate and lactylation in cancer. Signal Transduct Target Ther 2025; 10:38. [PMID: 39934144 DOI: 10.1038/s41392-024-02082-x] [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: 06/28/2024] [Revised: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 02/13/2025] Open
Abstract
Accumulated evidence has implicated the diverse and substantial influence of lactate on cellular differentiation and fate regulation in physiological and pathological settings, particularly in intricate conditions such as cancer. Specifically, lactate has been demonstrated to be pivotal in molding the tumor microenvironment (TME) through its effects on different cell populations. Within tumor cells, lactate impacts cell signaling pathways, augments the lactate shuttle process, boosts resistance to oxidative stress, and contributes to lactylation. In various cellular populations, the interplay between lactate and immune cells governs processes such as cell differentiation, immune response, immune surveillance, and treatment effectiveness. Furthermore, communication between lactate and stromal/endothelial cells supports basal membrane (BM) remodeling, epithelial-mesenchymal transitions (EMT), metabolic reprogramming, angiogenesis, and drug resistance. Focusing on lactate production and transport, specifically through lactate dehydrogenase (LDH) and monocarboxylate transporters (MCT), has shown promise in the treatment of cancer. Inhibitors targeting LDH and MCT act as both tumor suppressors and enhancers of immunotherapy, leading to a synergistic therapeutic effect when combined with immunotherapy. The review underscores the importance of lactate in tumor progression and provides valuable perspectives on potential therapeutic approaches that target the vulnerability of lactate metabolism, highlighting the Heel of Achilles for cancer treatment.
Collapse
Affiliation(s)
- Jie Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ziyue Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ya Chen
- Department of Radiology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Hao Tian
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Yongning Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Shiqiong Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| |
Collapse
|
21
|
Pham DX, Hsu T. Tumor-initiating and metastasis-initiating cells of clear-cell renal cell carcinoma. J Biomed Sci 2025; 32:17. [PMID: 39920694 PMCID: PMC11806631 DOI: 10.1186/s12929-024-01111-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/11/2024] [Indexed: 02/09/2025] Open
Abstract
Clear-cell renal cell carcinoma (ccRCC) is the most common subtype of kidney malignancy. ccRCC is considered a major health concern worldwide because its numbers of incidences and deaths continue to rise and are predicted to continue rising in the foreseeable future. Therefore new strategy for early diagnosis and therapeutics for this disease is urgently needed. The discovery of cancer stem cells (CSCs) offers hope for early cancer detection and treatment. However, there has been no definitive identification of these cancer progenitors for ccRCC. A majority of ccRCC is characterized by the loss of the von Hippel-Lindau (VHL) tumor suppressor gene function. Recent advances in genome analyses of ccRCC indicate that in ccRCC, tumor-initiating cells (TICs) and metastasis-initiating cells (MICs) are two distinct groups of progenitors. MICs result from various genetic changes during subclonal evolution, while TICs reside in the stem of the ccRCC phylogenetic tree of clonal development. TICs likely originate from kidney tubule progenitor cells bearing VHL gene inactivation, including chromatin 3p loss. Recent studies also point to the importance of microenvironment reconstituted by the VHL-deficient kidney tubule cells in promoting ccRCC initiation and progression. These understandings should help define the progenitors of ccRCC and facilitate early detection and treatment of this disease.
Collapse
Affiliation(s)
- Dinh-Xuan Pham
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan, ROC
| | - Tien Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan, ROC.
- Graduate Institute of Biomedical Sciences, China Medical University-Taiwan, No. 91 Hsueh-Shih Road, Taichung, 40402, Taiwan, ROC.
| |
Collapse
|
22
|
Yao ZY, Ma X, Cui YZ, Liu J, Han ZX, Song J. Impact of triglyceride-glucose index on the long-term prognosis of advanced gastric cancer patients receiving immunotherapy combined with chemotherapy. World J Gastroenterol 2025; 31:102249. [PMID: 39926212 PMCID: PMC11718607 DOI: 10.3748/wjg.v31.i5.102249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/09/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the fifth most common malignancy and the third leading cause of death worldwide. Despite advancements in immunotherapies, patient prognosis remains poor, necessitating the identification of key prognostic factors to optimize the treatment approaches. Insulin resistance, as indicated by the triglyceride glucose (TyG) index, is increasingly recognized for its impact on cancer progression and immune modulation, and its potential role in GC prognosis is of particular interest. AIM To investigate whether the TyG index, a surrogate marker of insulin resistance, can predict the prognosis of patients with advanced GC receiving immunotherapy combined with chemotherapy. METHODS This retrospective study included 300 patients with advanced GC who received sintilimab combined with chemotherapy. The patients were categorized into two groups according to high or low TyG index, and independent prognostic factors for overall survival (OS) were determined using Cox proportional hazards regression analysis, which led to the development of a nomogram model. RESULTS Of the included patients, 136 had a high TyG index and 164 had a low TyG index. The median progression-free survival of the high TyG index group was significantly longer than that of the low TyG index group. Similarly, the median OS of the high TyG index group was significantly longer than that of the low TyG index group. The objective response and disease control rates in the two groups were 18.38% vs 9.15% and 58.82% vs 46.95%, respectively. No significant difference was noted in the incidence of adverse reactions at any level between the two groups (P > 0.05). In multivariate analysis, the Eastern Cooperative Oncology Group score, programmed cell death ligand 1 expression, and TyG index acted as independent prognostic factors for OS. Of these factors, the hazard ratio of the TyG index was 0.36 (95% confidence interval: 0.36-0.55, P < 0.001), and the nomogram model re-emphasized its importance as the main predictor of patient prognosis, followed by programmed cell death ligand 1 expression and the Eastern Cooperative Oncology Group score. CONCLUSION The TyG index is a long-term predictor of the efficacy of immunotherapy combined with chemotherapy, and patients with a high index have a better prognosis.
Collapse
Affiliation(s)
- Zhi-Yuan Yao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
| | - Xiao Ma
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Yong-Zheng Cui
- Department of Radiotherapy, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
| | - Jie Liu
- Department of Radiotherapy, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
| | - Zheng-Xiang Han
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
| | - Jun Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, Jiangsu Province, China
| |
Collapse
|
23
|
Kim J, Seki E. Inflammation and Immunity in Liver Neoplasms: Implications for Future Therapeutic Strategies. Mol Cancer Ther 2025; 24:188-199. [PMID: 39365846 PMCID: PMC11794036 DOI: 10.1158/1535-7163.mct-23-0726] [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: 06/04/2024] [Revised: 07/28/2024] [Accepted: 08/09/2024] [Indexed: 10/06/2024]
Abstract
Over the past two decades, the "hallmarks of cancer" have revolutionized cancer research and highlighted the crucial roles of inflammation and immunity. Protumorigenic inflammation promotes cancer development along with inhibition of antitumor immunity, shaping the tumor microenvironment (TME) toward a tumor-permissive state and further enhancing the malignant potential of cancer cells. This immunosuppressive TME allows tumors to evade immunosurveillance. Thus, understanding the complex interplay between tumors and the immune system within the TME has become pivotal, especially with the advent of immunotherapy. Although immunotherapy has achieved notable success in many malignancies, primary liver cancer, particularly hepatocellular carcinoma, presents unique challenges. The hepatic immunosuppressive environment poses obstacles to the effectiveness of immunotherapy, along with high mortality rates and limited treatment options for patients with liver cancer. In this review, we discuss current understanding of the complex immune-mediated mechanisms underlying liver neoplasms, focusing on hepatocellular carcinoma and liver metastases. We describe the molecular and cellular heterogeneity within the TME, highlighting how this presents unique challenges and opportunities for immunotherapy in liver cancers. By unraveling the immune landscape of liver neoplasms, this review aims to contribute to the development of more effective therapeutic interventions, ultimately improving clinical outcomes for patients with liver cancer.
Collapse
Affiliation(s)
- Jieun Kim
- Karsh Division of Gastroenterology Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
24
|
Doi H, Muraguchi H, Horio T, Choi YJ, Takahashi K, Noda T, Sawada K. Real-time simultaneous visualization of lactate and proton dynamics using a 6-μm-pitch CMOS multichemical image sensor. Biosens Bioelectron 2025; 268:116898. [PMID: 39522470 DOI: 10.1016/j.bios.2024.116898] [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: 03/19/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Multi-analyte detection and imaging of extracellular chemical signaling molecules are crucial for understanding brain function and molecular pathology. In this work, we present a 6-μm-pitch, CMOS-based multichemical image sensor that enables the simultaneous visualization and spatiotemporal multimodal analysis of the lactate and proton (H+) dynamics without any labeling. Using semiconductor lithography, gold electrode patterns functioning as lactate-sensing regions were formed on a potentiometric sensor array. Lactate is detected potentiometrically because of redox reactions using lactate oxidase and horseradish peroxidase. The resulting multichemical image sensor exhibited a pH sensitivity of 65 mV and a superior detection limit of 1 μM for lactate with a reasonable selectivity. Furthermore, diffusion images of lactate and H+ distributions were obtained concurrently, allowing for simultaneous real-time imaging of the two chemicals with subcellular resolution. We believe that our novel imaging device can be successfully applied to extracellular microenvironments in tissue or cell samples as an effective bioimaging tool.
Collapse
Affiliation(s)
- Hideo Doi
- Electrical and Electronic Information Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempakucho, Toyohashi, Aichi, 441-8122, Japan.
| | - Hayato Muraguchi
- Electrical and Electronic Information Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempakucho, Toyohashi, Aichi, 441-8122, Japan
| | - Tomoko Horio
- Electrical and Electronic Information Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempakucho, Toyohashi, Aichi, 441-8122, Japan
| | - Yong-Joon Choi
- Electrical and Electronic Information Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempakucho, Toyohashi, Aichi, 441-8122, Japan
| | - Kazuhiro Takahashi
- Electrical and Electronic Information Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempakucho, Toyohashi, Aichi, 441-8122, Japan; Institute for Research on Next-generation Semiconductor and Sensing Science (IRES(2)), Toyohashi University of Technology, 1-1 Hibarigaoka, Tempakucho, Toyohashi, Aichi, 441-8122, Japan
| | - Toshihiko Noda
- Electrical and Electronic Information Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempakucho, Toyohashi, Aichi, 441-8122, Japan; Institute for Research on Next-generation Semiconductor and Sensing Science (IRES(2)), Toyohashi University of Technology, 1-1 Hibarigaoka, Tempakucho, Toyohashi, Aichi, 441-8122, Japan
| | - Kazuaki Sawada
- Electrical and Electronic Information Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempakucho, Toyohashi, Aichi, 441-8122, Japan; Institute for Research on Next-generation Semiconductor and Sensing Science (IRES(2)), Toyohashi University of Technology, 1-1 Hibarigaoka, Tempakucho, Toyohashi, Aichi, 441-8122, Japan
| |
Collapse
|
25
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
26
|
Pang B, Liu Z, Gao Y, Li X, Wang S, Qi M, Zhao X, Fan R, Xu D, Cullen PJ, Zhou R. Enhanced Anticancer Efficacy of Alkaline Plasma-Activated Water through Augmented RONS Production. ACS APPLIED MATERIALS & INTERFACES 2025; 17:467-483. [PMID: 39692225 DOI: 10.1021/acsami.4c16518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Despite notable advances in anticancer drug development, their manufacture and use pose environmental and health risks due to toxic byproducts, drug residue contamination, and cytotoxicity to normal cells. Therefore, developing cost-effective anticancer treatments with fewer toxic side effects and higher selectivity is essential to the advancement of highly effective anticancer therapies. Plasma-activated water (PAW) offers a green alternative to conventional chemical treatments as it reverts to water within days. However, the limited duration and dose of reactive oxygen and nitrogen species (RONS) in acidified PAW restrict its clinical deployment and the full understanding of their mechanism. In this study, we propose alkaline PAW as an innovative enhancement of the RONS technology. The alkaline PAW generated markedly superior RONS, with about 10 times higher levels of NO2-, H2O2, and ONOO-/O2•- than acidic PAW. The possible RONS generation pathways in alkaline PAW are analyzed by scavengers. In conventional acidic PAW, 70% of the H2O2 concentration is contributed by •OH but only about 20% in alkaline PAW. ONOO- is mainly formed through the reaction of O2•- with NO in alkaline pH, while in acidic PAW, it mainly forms from NO2- and H2O2. The results unveiled the synergistic and formidable anticancer effects of alkaline PAW against cancer cells, typified by an increase in intracellular ROS/RNS levels. Furthermore, alkaline PAW injection also effectively prevented xenograft tumor growth in mice. We systematically investigated this high-dose anticancer solution without using noble gases, toxic reagents, or extra energy consumption and successfully demonstrated the possibility of alkaline PAW being an effective and environmentally friendly therapeutic technology. The activity is closely linked to the RONS dose, and the generation pathway provides much-needed insight into the fundamental aspects of PAW chemistry required for the optimization of the biochemical activity of PAW.
Collapse
Affiliation(s)
- Bolun Pang
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Zhijie Liu
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Yuting Gao
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Xin Li
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Sitao Wang
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Miao Qi
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Xinyi Zhao
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Runze Fan
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Dehui Xu
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Patrick J Cullen
- School of Chemical and Biomolecular Engineering, University of Sydney, Sydney, NSW 2006, Australia
| | - Renwu Zhou
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| |
Collapse
|
27
|
Zhao W, Kim B, Coffey NJ, Bowers S, Jiang Y, Bowman CE, Noji M, Jang C, Simon MC, Arany Z, Kim B. HIF2α inhibits glutaminase clustering in mitochondria to sustain growth of clear cell Renal Cell Carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.04.592520. [PMID: 38746132 PMCID: PMC11092754 DOI: 10.1101/2024.05.04.592520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Clear cell renal cell carcinomas (ccRCC) are largely driven by HIF2α and are avid consumers of glutamine. However, inhibitors of glutaminase1 (GLS1), the first step in glutaminolysis, have not shown benefit in phase III trials, and HIF2α inhibition, recently FDA-approved for treatment of ccRCC, shows great but incomplete benefits, underscoring the need to better understand the roles of glutamine and HIF2α in ccRCC. Here, we report that glutamine deprivation rapidly redistributes GLS1 into isolated clusters within mitochondria across diverse cell types, but not in ccRCC. GLS1 clustering is rapid (1-3 hours) and reversible, is specifically driven by reduced intracellular glutamate, and is mediated by mitochondrial fission. Clustered GLS1 markedly enhances glutaminase activity and promotes cell death under glutamine-deprived conditions. HIF2α prevents GLS1 clustering, independently of its transcriptional activity, thereby protecting ccRCC cells from cell death induced by glutamine deprivation. Reversing this protection, by genetic expression of GLS1 mutants that constitutively cluster, enhances ccRCC cell death in culture and suppresses ccRCC growth in vivo. These findings provide multiple insights into cellular glutamine handling, including a novel metabolic pathway by which HIF2α promotes ccRCC, and reveals a potential therapeutic avenue to synergize with HIF2α inhibition in the treatment of ccRCC.
Collapse
Affiliation(s)
- Wencao Zhao
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boyoung Kim
- McAllister Heart Institute, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Nathan J Coffey
- The Abramson Family Cancer Research Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Schuyler Bowers
- McAllister Heart Institute, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yanqing Jiang
- The Abramson Family Cancer Research Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Michael Noji
- The Abramson Family Cancer Research Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, USA
| | - M. Celeste Simon
- The Abramson Family Cancer Research Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zoltan Arany
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boa Kim
- Department of Pathology and Laboratory Medicine, McAllister Heart Institute, Nutrition Obesity Research Center, and Lineberger Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
28
|
Torang A, Kirov AB, Lammers V, Cameron K, Wouters VM, Jackstadt RF, Lannagan TRM, de Jong JH, Koster J, Sansom O, Medema JP. Enterocyte-like differentiation defines metabolic gene signatures of CMS3 colorectal cancers and provides therapeutic vulnerability. Nat Commun 2025; 16:264. [PMID: 39747069 PMCID: PMC11696116 DOI: 10.1038/s41467-024-55574-3] [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: 04/13/2023] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Colorectal cancer (CRC) is stratified into four consensus molecular subtypes (CMS1-4). CMS3 represents the metabolic subtype, but its wiring remains largely undefined. To identify the underlying tumorigenesis of CMS3, organoids derived from 16 genetically engineered mouse models are analyzed. Upon in vitro Cre-recombinase activation, transformation is established and transcriptional profiling reveals that distinct CMSs (CMS2-4) are modeled with different organoids. CMS3-like, metabolic signature-positive, organoids are induced by KRAS mutations. Interestingly, metabolic signatures are subsequently shown to result from enterocyte-like differentiation both in organoids and human cancers. Further analysis reveals carbamoyl-phosphate synthase 1 (CPS1) and sucrase-isomaltase (SI) as signature proteins. More importantly, CPS1 is crucial for de novo pyrimidine synthesis in CMS3 and its inhibition targets proliferation and stemness, facilitating enterocyte-like differentiation, while CMS2 and CMS4 models are not affected. Our data point to an enterocyte-like differentiation of CMS3 CRCs and reveal a selective vulnerability of this subtype through CPS1 inhibition.
Collapse
Affiliation(s)
- Arezo Torang
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
| | - Aleksandar B Kirov
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
| | - Veerle Lammers
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
| | - Kate Cameron
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
| | - Valérie M Wouters
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
| | - Rene F Jackstadt
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Heidelberg, Germany Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | | | - Joan H de Jong
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
| | - Jan Koster
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Owen Sansom
- Cancer Research UK Scotland Institute, Garscube Estate, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
- Oncode Institute, Amsterdam, the Netherlands.
| |
Collapse
|
29
|
Liu J, Li X, Li Y, Gong Q, Luo K. Metformin-based nanomedicines for reprogramming tumor immune microenvironment. Theranostics 2025; 15:993-1016. [PMID: 39776799 PMCID: PMC11700864 DOI: 10.7150/thno.104872] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
Immunotherapy has transformed current cancer management, and it has achieved significant progress over last decades. However, an immunosuppressive tumor microenvironment (TME) diminishes the effectiveness of immunotherapy by suppressing the activity of immune cells and facilitating tumor immune-evasion. Adenosine monophosphate-activated protein kinase (AMPK), a key modulator of cellular energy metabolism and homeostasis, has gained growing attention in anti-tumor immunity. Metformin is usually considered as a cornerstone in diabetes management, and its role in activating the AMPK pathway has also been extensively explored in cancer therapy although the findings on its role remain inconsistent. Metformin in a nanomedicine formulation has been found to hold potential in reprogramming the immunosuppressive TME through immunometabolic modulation of both tumor and immune cells. This review elaborates the foundation and progress of immunometabolic reprogramming of the TME via metformin-based nanomedicines, offering valuable insights for the next generation of cancer therapy.
Collapse
Affiliation(s)
- Jieyu Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoling Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinggang Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, NHC Key Laboratory of Transplant Engineering and Immunology, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Xiamen Key Lab of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, NHC Key Laboratory of Transplant Engineering and Immunology, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
30
|
Anandi L, Garcia J, Ros M, Janská L, Liu J, Carmona-Fontaine C. Direct visualization of emergent metastatic features within an ex vivo model of the tumor microenvironment. Life Sci Alliance 2025; 8:e202403053. [PMID: 39419548 PMCID: PMC11487089 DOI: 10.26508/lsa.202403053] [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: 09/19/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Ischemic conditions such as hypoxia and nutrient starvation, together with interactions with stromal cells, are critical drivers of metastasis. These conditions arise deep within tumor tissues, and thus, observing nascent metastases is exceedingly challenging. We thus developed the 3MIC-an ex vivo model of the tumor microenvironment-to study the emergence of metastatic features in tumor cells in a 3-dimensional (3D) context. Here, tumor cells spontaneously create ischemic-like conditions, allowing us to study how tumor spheroids migrate, invade, and interact with stromal cells under different metabolic conditions. Consistent with previous data, we show that ischemia increases cell migration and invasion, but the 3MIC allowed us to directly observe and perturb cells while they acquire these pro-metastatic features. Interestingly, our results indicate that medium acidification is one of the strongest pro-metastatic cues and also illustrate using the 3MIC to test anti-metastatic drugs on cells experiencing different metabolic conditions. Overall, the 3MIC can help dissecting the complexity of the tumor microenvironment for the direct observation and perturbation of tumor cells during the early metastatic process.
Collapse
Affiliation(s)
- Libi Anandi
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Jeremy Garcia
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Manon Ros
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Libuše Janská
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Josephine Liu
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Carlos Carmona-Fontaine
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| |
Collapse
|
31
|
Yang L, Li A, Yu W, Wang H, Zhang L, Wang D, Wang Y, Zhang R, Lei Q, Liu Z, Zhen S, Qin H, Liu Y, Yang Y, Song XL, Zhang Y. Blockade of purine metabolism reverses macrophage immunosuppression and enhances anti-tumor immunity in non-small cell lung cancer. Drug Resist Updat 2025; 78:101175. [PMID: 39608215 DOI: 10.1016/j.drup.2024.101175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/17/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
AIMS Immune checkpoint blockade therapy is not effective in most patients with non-small cell lung cancer (NSCLC) due to the immunosuppressive tumor microenvironment. Macrophages are key components of tumor-infiltrating immune cells and play a critical role in immunosuppression, which can be mediated by cell-intrinsic metabolism. This study aimed to evaluate whether macrophages regulate NSCLC progression through metabolic crosstalk with cancer cells and affect immunotherapy efficacy. METHODS The macrophage landscape of NSCLC tissues were analyzed by single-cell sequencing and verified through flow cytometry and immunofluorescence. Multiplex assay, single-cell sequencing data, ELISA, immunofluorescence, and RNA-seq et al. were used to investigate and verify the mechanism of macrophage-mediated metabolic regulation on immunosuppression. The tumor-bearing model was established in C57BL/6 J mice to explore in vivo efficacy. RESULTS We found that tumor tissue-derived macrophages exhibited an anti-inflammatory phenotype and had a prognostic value for NSCLC. NSCLC cell-secreted CXCL8 recruited macrophages from peritumor tissues to tumor sites and promoted programmed death-ligand 1 (PD-L1) expression by activating purine metabolism with increasing xanthine dehydrogenase and uric acid production. Moreover, purine metabolism-mediated macrophage immunosuppression was dependent on NLRP3/caspase-1/IL-1β signaling. Blockade of purine metabolism signaling enhanced anti-tumor immunity and the efficacy of anti-PD-L1 therapy. CONCLUSIONS Collectively, our findings reveal a key role of purine metabolism in macrophage immunosuppression and suggest that blockade of purine metabolism combined with immune checkpoint blockade could provide synergistic effects in NSCLC treatment.
Collapse
Affiliation(s)
- Li Yang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China; School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Aitian Li
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Weina Yu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huishang Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lei Zhang
- Thoracic Surgery Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dan Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ru Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingyang Lei
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhangnan Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shanshan Zhen
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haiming Qin
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yaqing Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Yang
- Thoracic Surgery Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xian-Lu Song
- Department of Radiotherapy, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China; School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Public Health, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
32
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
33
|
Shafi S, Khan MA, Ahmad J, Rabbani SA, Singh S, Najmi AK. Envisioning Glucose Transporters (GLUTs and SGLTs) as Novel Intervention against Cancer: Drug Discovery Perspective and Targeting Approach. Curr Drug Targets 2025; 26:109-131. [PMID: 39377414 DOI: 10.2174/0113894501335877240926101134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 10/09/2024]
Abstract
Metabolic reprogramming and altered cellular energetics have been recently established as an important cancer hallmark. The modulation of glucose metabolism is one of the important characteristic features of metabolic reprogramming in cancer. It contributes to oncogenic progression by supporting the increased biosynthetic and bio-energetic demands of tumor cells. This oncogenic transformation consequently results in elevated expression of glucose transporters in these cells. Moreover, various cancers exhibit abnormal transporter expression patterns compared to normal tissues. Recent investigations have underlined the significance of glucose transporters in regulating cancer cell survival, proliferation, and metastasis. Abnormal regulation of these transporters, which exhibit varying affinities for hexoses, could enable cancer cells to efficiently manage their energy supply, offering a crucial edge for proliferation. Exploiting the upregulated expression of glucose transporters, GLUTs, and Sodium Linked Glucose Transporters (SGLTs), could serve as a novel therapeutic intervention for anti-cancer drug discovery as well as provide a unique targeting approach for drug delivery to specific tumor tissues. This review aims to discussthe previous and emerging research on the expression of various types of glucose transporters in tumor tissues, the role of glucose transport inhibitors as a cancer therapy intervention as well as emerging GLUT/SGLT-mediated drug delivery strategies that can be therapeutically employed to target various cancers.
Collapse
Affiliation(s)
- Sadat Shafi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Kingdom of Saudi Arabia (KSA)
| | - Syed Arman Rabbani
- Department of Clinical Pharmacy and Pharmacology, Ras Al Khaimah College of Pharmacy, Ras Al Khaimah Medical and Health Science University, Ras Al Khaimah, United Arab Emirates
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
34
|
Ahmad I, Altameemi KKA, Hani MM, Ali AM, Shareef HK, Hassan ZF, Alubiady MHS, Al-Abdeen SHZ, Shakier HG, Redhee AH. Shifting cold to hot tumors by nanoparticle-loaded drugs and products. Clin Transl Oncol 2025; 27:42-69. [PMID: 38922537 DOI: 10.1007/s12094-024-03577-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Cold tumors lack antitumor immunity and are resistant to therapy, representing a major challenge in cancer medicine. Because of the immunosuppressive spirit of the tumor microenvironment (TME), this form of tumor has a low response to immunotherapy, radiotherapy, and also chemotherapy. Cold tumors have low infiltration of immune cells and a high expression of co-inhibitory molecules, such as immune checkpoints and immunosuppressive molecules. Therefore, targeting TME and remodeling immunity in cold tumors can improve the chance of tumor repression after therapy. However, tumor stroma prevents the infiltration of inflammatory cells and hinders the penetration of diverse molecules and drugs. Nanoparticles are an intriguing tool for the delivery of immune modulatory agents and shifting cold to hot tumors. In this review article, we discuss the mechanisms underlying the ability of nanoparticles loaded with different drugs and products to modulate TME and enhance immune cell infiltration. We also focus on newest progresses in the design and development of nanoparticle-based strategies for changing cold to hot tumors. These include the use of nanoparticles for targeted delivery of immunomodulatory agents, such as cytokines, small molecules, and checkpoint inhibitors, and for co-delivery of chemotherapy drugs and immunomodulatory agents. Furthermore, we discuss the potential of nanoparticles for enhancing the efficacy of cancer vaccines and cell therapy for overcoming resistance to treatment.
Collapse
Affiliation(s)
- Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia.
| | | | - Mohaned Mohammed Hani
- Department of Medical Instrumentation Engineering Techniques, Imam Ja'afar Al-Sadiq University, Al Muthanna, Iraq
| | - Afaq Mahdi Ali
- Department of Pharmaceutics, Al-Turath University College, Baghdad, Iraq
| | - Hasanain Khaleel Shareef
- Department of Medical Biotechnology, College of Science, Al-Mustaqbal University, Hilla, Iraq
- Biology Department, College of Science for Women, University of Babylon, Hilla, Iraq
| | | | | | | | | | - Ahmed Huseen Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
35
|
Sharma NK, Sarode SC, Sekar G, Sonawane K, Bomle D. Challenges in Metabolite Biomarkers as Avenues of Diagnosis and Prognosis of Cancer. J Cancer Prev 2024; 29:105-112. [PMID: 39790219 PMCID: PMC11706722 DOI: 10.15430/jcp.24.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/09/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025] Open
Abstract
Given the evolutionary nature of tumor complexities and heterogeneity, the early diagnosis of cancer encounters various challenges. Complexities at the level of metabolite reprogramming are compelling in the background of invasiveness, metastasis, drug- and radiation-induced metabolic alterations, immunotherapy-influenced changes, and pro-tumor niche including microbiome. Therefore, it is crucial to examine both current and future obstacles associated with early cancer detection specifically in the context of tumor metabolite biomarkers at preclinical and clinical levels. In conclusion, the significance of tumor metabolite biomarkers must be aligned with a comprehensive approach to achbieve diagnosis and prognosis of cancer patients by securing solutions to formidable challenges.
Collapse
Affiliation(s)
- Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
| | - Sachin C. Sarode
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, India
| | - Gopinath Sekar
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
- Department of Biotechnology, Sri Venkateswara College of Engineering, Sriperumbudur, India
| | - Kaveri Sonawane
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
| | - Dhanashree Bomle
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
| |
Collapse
|
36
|
Diao B, Fan Z, Zhou B, Zhan H. Crosstalk between pancreatic cancer and adipose tissue: Molecular mechanisms and therapeutic implications. Biochem Biophys Res Commun 2024; 740:151012. [PMID: 39561650 DOI: 10.1016/j.bbrc.2024.151012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
The incidence rate of pancreatic cancer, a fatal illness with a meager 5-year survival rate, has been on the rise in recent times. When individuals accumulate excessive amounts of adipose tissue, the adipose organ becomes dysfunctional due to alterations in the adipose tissue microenvironment associated with inflammation and metabolism. This phenomenon may potentially contribute to the aberrant accumulation of fat that initiates pancreatic carcinogenesis, thereby influencing the disease's progression, resistance to treatment, and metastasis. This review presents a summary of the impact of pancreatic steatosis, visceral fat, cancer-associated adipocytes and lipid diets on the advancement of pancreatic cancer, as well as the reciprocal effects of pancreatic cancer on adipose tissue. Understanding the molecular mechanisms underlying the relationship between dysfunctional adipose tissue and pancreatic cancer better may lead to the discovery of new therapeutic targets for the disease's prevention and individualized treatment. This is especially important given the rising global incidence of obesity, which will improve the pancreatic cancer treatment options that are currently insufficient.
Collapse
Affiliation(s)
- Boyu Diao
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
37
|
He J, Du M, Bi X, Chen P, Li J, Tian R, Fan L, Zhang Q. Construction and evaluation of a prognostic model for metabolism-related genes in kidney renal clear cell carcinoma using TCGA database. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2024; 12:352-366. [PMID: 39839750 PMCID: PMC11744353 DOI: 10.62347/xvzj5704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/15/2024] [Indexed: 01/23/2025]
Abstract
OBJECTIVE To investigate the expression of metabolism-related genes (MRGs) in kidney renal clear cell carcinoma (KIRC) and their association with patient prognosis, and to identify potential targets for intervention. METHODS Bioinformatics methods were employed to mine the KIRC transcriptome data in The Cancer Genome Atlas Program (TCGA) database in order to identify MRGs that are aberrantly expressed in cancerous tissues. Subsequently, a prognostic risk score model was constructed and its predictive capacity was evaluated. Finally, the expression of prognostically relevant MRGs was validated using external datasets and KIRC clinical samples. RESULTS A total of 789 differentially expressed MRGs associated with KIRC were screened, of which 465 genes were upregulated and 324 genes were downregulated, and finally 23 genes were screened to establish a risk score model. We found that the AUCs of the risk score model for predicting patients' 1-, 3- and 5-year overall survival (OS) were 0.804, 0.766 and 0.802, respectively. These findings suggest that the model has good predictive ability. A multifactorial Cox analysis revealed that 23 MRGs risk score was significantly associated with the overall survival of KIRC patients, and could therefore be used as an independent risk factor for the prognosis of KIRC patients (HR = 3.495, P < 0.001). Meanwhile, Kaplan-Meier analyses of the high-risk and low-risk groups indicated that the high-risk group exhibited a markedly inferior overall survival (OS) prognosis. The validation of clinical samples from KIRC patients and four external data sets (GSE36895, GSE40435, GSE53757 and GSE66272) demonstrated that KCNN4 and PLK1 were highly expressed in KIRC, whereas TEK, PLG, ANGPTL3, TFAP2A, ANK3, ATP1A1 and UCN exhibited low expression in KIRC. CONCLUSION Several MRGs are aberrantly expressed in KIRC, from which we screened 23 genes and constructed a MRGs prognostic risk model that can effectively predict the prognosis of KIRC patients and provide a new foundation for personalised diagnosis and treatment of KIRC.
Collapse
Affiliation(s)
- Jingteng He
- Department of Urology, General Hospital of Northern Theater CommandShenyang 110016, Liaoning, China
| | - Mou Du
- Department of Urology, General Hospital of Northern Theater CommandShenyang 110016, Liaoning, China
| | - Xiaojun Bi
- Department of Urology, General Hospital of Northern Theater CommandShenyang 110016, Liaoning, China
| | - Peng Chen
- Department of Urology, General Hospital of Northern Theater CommandShenyang 110016, Liaoning, China
| | - Jian Li
- Department of Urology, General Hospital of Northern Theater CommandShenyang 110016, Liaoning, China
| | - Renli Tian
- Department of Urology, General Hospital of Northern Theater CommandShenyang 110016, Liaoning, China
| | | | | |
Collapse
|
38
|
Nelson AB, Reese LE, Rono E, Queathem ED, Qiu Y, McCluskey BM, Crampton A, Conniff E, Cummins K, Boytim E, Dansou S, Hwang J, Safo S, Puchalska P, Wood DK, Schwertfeger KL, Crawford PA. Deciphering Colorectal Cancer-Hepatocyte Interactions: A Multiomic Platform for Interrogation of Metabolic Crosstalk in the Liver-Tumor Microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627264. [PMID: 39713297 PMCID: PMC11661097 DOI: 10.1101/2024.12.06.627264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to adapt to and exploit their microenvironment for sustained growth. The liver is a common site of metastasis, but the interactions between tumor cells and hepatocytes remain poorly understood. In the context of liver metastasis, these interactions play a crucial role in promoting tumor survival and progression. This study leverages multiomics coverage of the microenvironment via liquid chromatography and high-resolution, high-mass accuracy mass spectrometry-based untargeted metabolomics, 13C-stable isotope tracing, and RNA sequencing to uncover the metabolic impact of co-localized primary hepatocytes and a colon adenocarcinoma cell line, SW480, using a 2D co-culture model. Metabolic profiling revealed disrupted Warburg metabolism with an 80% decrease in glucose consumption and 94% decrease in lactate production by hepatocyte-SW480 co-cultures relative to SW480 control cultures. Decreased glucose consumption was coupled with alterations in glutamine and ketone body metabolism, suggesting a possible fuel switch upon co-culturing. Further, integrated multiomic analysis indicates that disruptions in metabolic pathways, including nucleoside biosynthesis, amino acids, and TCA cycle, correlate with altered SW480 transcriptional profiles and highlight the importance of redox homeostasis in tumor adaptation. Finally, these findings were replicated in 3-dimensional microtissue organoids. Taken together, these studies support a bioinformatic approach to study metabolic crosstalk and discovery of potential therapeutic targets in preclinical models of the tumor microenvironment.
Collapse
Affiliation(s)
- Alisa B. Nelson
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
| | - Lyndsay E. Reese
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN USA
| | - Elizabeth Rono
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Eric D. Queathem
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN USA
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Yinjie Qiu
- Minnesota Supercomputing Institute, Minneapolis, MN USA
| | | | - Alexandra Crampton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Eric Conniff
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Katherine Cummins
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Ella Boytim
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Senali Dansou
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Justin Hwang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Sandra Safo
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, MN, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
| | - David K. Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Kathryn L. Schwertfeger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| |
Collapse
|
39
|
Wu B, Wang Z, Liu J, Li N, Wang X, Bai H, Wang C, Shi J, Zhang S, Song J, Li Y, Nie G. Dual rectification of metabolism abnormality in pancreatic cancer by a programmed nanomedicine. Nat Commun 2024; 15:10526. [PMID: 39627234 PMCID: PMC11615375 DOI: 10.1038/s41467-024-54963-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive and lethal malignancy characterized by dysregulated energy and stromal metabolism. It is strongly supported by activated pancreatic stellate cells (PSC) which drive excessive desmoplasia and tumor growth via metabolic crosstalk. Herein, a programmed nanosystem is designed to dual rectify the metabolism abnormalities of the PDAC cells, which overexpress glucose transporter 1(GLUT1) and CD71, and the PSC for oncotherapy. The nanosystem is based on a tumor microenvironment-responsive liposome encapsulating an NF-κB inhibitor (TPCA-1) and a CD71 aptamer-linked Glut1 siRNA. TPCA-1 reverses the activated PSC to quiescence, which hampers metabolic support of the PSC to PDAC cells and bolsters the PDAC cell-targeting delivery of the siRNA. Aerobic glycolysis and the following enhancement of oxidative phosphorylation are restrained by the nano-modulation so as to amplify anti-PDAC efficacy in an orthotopic xenograft mouse model, which implies more personalized PDAC treatment based on different energy metabolic profiles.
Collapse
MESH Headings
- Animals
- Humans
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Cell Line, Tumor
- Mice
- Nanomedicine/methods
- Liposomes/metabolism
- Pancreatic Stellate Cells/metabolism
- Pancreatic Stellate Cells/pathology
- Tumor Microenvironment
- Glucose Transporter Type 1/metabolism
- Glucose Transporter Type 1/genetics
- RNA, Small Interfering/metabolism
- RNA, Small Interfering/genetics
- NF-kappa B/metabolism
- Xenograft Model Antitumor Assays
- Receptors, Transferrin/metabolism
- Receptors, Transferrin/genetics
- Oxidative Phosphorylation
- Glycolysis
- Mice, Nude
- Aptamers, Nucleotide/metabolism
Collapse
Affiliation(s)
- Bowen Wu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
- Henan Institute of Advanced Technology, Henan, PR China
| | - Zhiqin Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
- College of Pharmaceutical Science, Jilin University, Changchun, PR China
| | - Jingyuan Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - Naishi Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - Xudong Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - HaoChen Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - Chunling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China
| | - Saiyang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Yiye Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China.
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, PR China.
| | - Guangjun Nie
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China.
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, PR China.
- Henan Institute of Advanced Technology, Henan, PR China.
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, PR China.
| |
Collapse
|
40
|
Jia Y, Wang F, Chen S, Wang J, Gao Y. Long-term hypoxia-induced physiological response in turbot Scophthalmus maximus L. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:2407-2421. [PMID: 39190213 DOI: 10.1007/s10695-024-01398-3] [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: 03/04/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024]
Abstract
Hypoxia affects fish's survival, growth, and physiological metabolism processes. In this study, turbot plasma glucose and cortisol contents, hepatic glycolysis (hexokinase [HK], phosphofructokinase [PFK], pyruvate kinase [PK]) and lipolysis (fatty acid synthetase [FAS], lipoprotein lipase [LPL]) enzyme activities, anti-oxidant enzyme (superoxide dismutase [SOD], catalase [CAT], glutathione peroxidase [GSH-Px]) activities, malondialdehyde (MDA), lactate and glycogen contents, gill histological parameters (lamellar length [SLL], width [SLW], interlamellar distance [ID]), respiratory frequency (RF), the proportion of the secondary lamellae available for gas exchange (PAGE), and hifs (hif-1α, hif-2α, hif-3α) expression were determined during long-term hypoxia and reoxygenation. Results showed that long-term hypoxia (3.34 ± 0.17 mg L-1) significantly elevated plasma cortisol and glucose contents; increased hepatic HK, PK, PFK, FAS, and LPL activity; decreased hepatic glycogen, lactate contents, and lipid drop numbers; and caused changes of hepatocyte (vacuolation, pyknotic, and lytic nucleus) after treatment for 4 weeks. Hepatic SOD, CAT, GSH-Px activity, and MDA contents; lamellar perimeter, SLL, ID, RF, and PAGE; and hepatic hif-1α, hif-2α, and hif-3α manifested similar results. Meanwhile, hif-1α is significantly higher than hif-2α, and hif-3α. Interestingly, females and males demonstrated no sex dimorphism significantly different from the above parameters (except hepatic FAS, LPL activity, and lipid drop number) under hypoxia. The above parameters recovered to normal levels after reoxygenation treatment for 4 weeks. Thus, long-term hypoxia promotes turbot hepatic glycogenolysis and lipolysis, induces oxidative damage and stimulates hepatic antioxidant capacity, and alters gill morphology to satisfy insufficient energy demand and alleviate potential damage, while hif-1α plays critical roles in the above physiological process.
Collapse
Affiliation(s)
- Yudong Jia
- Yellow Sea Fisheries Research Institute, Qingdao Key Laboratory for Marine Fish Breeding and Biotechnology, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China.
| | - Feng Wang
- Yellow Sea Fisheries Research Institute, Qingdao Key Laboratory for Marine Fish Breeding and Biotechnology, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Shuaiyu Chen
- Yellow Sea Fisheries Research Institute, Qingdao Key Laboratory for Marine Fish Breeding and Biotechnology, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Jiawei Wang
- Yellow Sea Fisheries Research Institute, Qingdao Key Laboratory for Marine Fish Breeding and Biotechnology, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China
| | - Yuntao Gao
- Yellow Sea Fisheries Research Institute, Qingdao Key Laboratory for Marine Fish Breeding and Biotechnology, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China
| |
Collapse
|
41
|
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
|
42
|
Liu H, Pan M, Liu M, Zeng L, Li Y, Huang Z, Guo C, Wang H. Lactate: a rising star in tumors and inflammation. Front Immunol 2024; 15:1496390. [PMID: 39660139 PMCID: PMC11628389 DOI: 10.3389/fimmu.2024.1496390] [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: 09/14/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Lactate has been traditionally regarded as a mere byproduct of glycolysis or metabolic waste. However, an increasing body of literature suggests its critical role in regulating various physiological and pathological processes. Lactate is generally associated with hypoxia, inflammation, viral infections, and tumors. It performs complex physiological roles by activating monocarboxylate transporter (MCT) or the G protein-coupled receptor GPR81 across the cell membrane. Lactate exerts immunosuppressive effects by regulating the functions of various immune cells (such as natural killer cells, T cells, dendritic cells, and monocytes) and its role in macrophage polarization and myeloid-derived suppressor cell (MDSC) differentiation in the tumor microenvironment. Lactic acid has also recently been found to increase the density of CD8+ T cells, thereby enhancing the antitumor immune response. Acute or chronic inflammatory diseases have opposite immune states in the inflammatory disease microenvironment. Factors such as cell types, transcriptional regulators, ionic mediators, and the microenvironment all contribute to the diverse functions lactate exhibits. Herein, we reviewed the pleiotropic effects of lactate on the regulation of various functions of immune cells in the tumor microenvironment and under inflammatory conditions, which may help to provide new insights and potential targets for the diagnosis and treatment of inflammatory diseases and malignancies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chunlei Guo
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
43
|
Weerasinghe HN, Burrage PM, Jr DVN, Burrage K. Agent-based modeling for the tumor microenvironment (TME). MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:7621-7647. [PMID: 39696854 DOI: 10.3934/mbe.2024335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Cancer is a disease that arises from the uncontrolled growth of abnormal (tumor) cells in an organ and their subsequent spread into other parts of the body. If tumor cells spread to surrounding tissues or other organs, then the disease is life-threatening due to limited treatment options. This work applies an agent-based model to investigate the effect of intra-tumoral communication on tumor progression, plasticity, and invasion, with results suggesting that cell-cell and cell-extracellular matrix (ECM) interactions affect tumor cell behavior. Additionally, the model suggests that low initial healthy cell densities and ECM protein densities promote tumor progression, cell motility, and invasion. Furthermore, high ECM breakdown probabilities of tumor cells promote tumor invasion. Understanding the intra-tumoral communication under cellular stress can potentially lead to the design of successful treatment strategies for cancer.
Collapse
Affiliation(s)
- Hasitha N Weerasinghe
- School of Mathematical Sciences, Queensland University of Technology, Queensland, Brisbane, Australia
| | - Pamela M Burrage
- School of Mathematical Sciences, Queensland University of Technology, Queensland, Brisbane, Australia
| | - Dan V Nicolau Jr
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Kevin Burrage
- School of Mathematical Sciences, Queensland University of Technology, Queensland, Brisbane, Australia
- Department of Computer Science, University of Oxford, United Kingdom
| |
Collapse
|
44
|
Li J, Zhai X, Chen C, Zhang R, Huang X, Liu Y. The intrahepatic bacterial metataxonomic signature of patients with hepatocellular carcinoma. Sci Rep 2024; 14:29077. [PMID: 39580523 PMCID: PMC11585554 DOI: 10.1038/s41598-024-80246-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024] Open
Abstract
Dysbiosis of the gut-liver axis increases the risk of bacterial and metabolite influx into the liver, which may contribute to the development of hepatocellular carcinoma (HCC). In this study, we compared the microbiomes in HCC tumors and adjacent tissues. We examined the HCC tumors and adjacent tissues from 19 patients diagnosed with HCC. We find that the liver tissues from HCC patients with capsule invasion presented higher alpha diversity at the genus level than those without. The bacterial compositions in liver tissues of HCC patients at stage II differed from those at stage I and Advanced, respectively. Metagenomic profiling revealed that order Actinomycetales was enriched in the HCC patients at advanced stages. Order Lactobacillales, family Veillonellaceae, genera Rhodobacter and Megasphaera are enriched in tumors of HCC patients, whereas genus Pseudochrobactrum is enriched in the adjacent tissues from HCC patients. An increased abundance of class Actinobacteria and order Actinomycetales is observed in the HCC patients with cirrhosis. In contrast, phylum Firmicutes, classes Clostridia and Betaproteobacteria, and order Clostridiales are enriched in those without cirrhosis. The presence of various types of bacterial 16S rRNAs in HCC tumors and adjacent tissues indicates the presence of various bacterial communities therein. Our study provides information about differentially abundant intrahepatic bacteria in patients with HCC. The differences found may support possible diagnostic and personalized therapeutic implications for HCC.
Collapse
Affiliation(s)
- Jie Li
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xuanpei Zhai
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, Shanghai, 201210, China
| | - Changzhou Chen
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai, China
| | - Rong Zhang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Xiaowu Huang
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.
| | - Yifan Liu
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, 201210, China.
- State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
45
|
Adham SA, Al Kalbani A, Al Zeheimi N, Al Dalali M, Al Kharusi N, Siddiqi A, Al Maskari A. Glycemic load impacts the response of acquired resistance in breast cancer cells to chemotherapeutic drugs in vitro. PLoS One 2024; 19:e0311345. [PMID: 39576770 PMCID: PMC11584130 DOI: 10.1371/journal.pone.0311345] [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: 03/26/2024] [Accepted: 09/17/2024] [Indexed: 11/24/2024] Open
Abstract
Resisting chemotherapy is a significant hurdle in treating breast cancer. Locally advanced breast cancer patients undergo four cycles of Adriamycin and Cyclophosphamide, followed by four cycles of Paclitaxel before surgery. Some patients resist this regimen, and their cancer recurred. Our study aimed to understand the underlying mechanisms of acquired resistance during these specific treatment phases. We explored how breast cancer cells, resistant to chemotherapy, respond to different glucose levels, shedding light on the intricate relationship between diabetes, breast cancer subtype, and resistance to preoperative chemotherapy. We examined two groups of cell lines: the standard MDA-MB-231 and MCF7 cells and their resistant counterparts after exposure to four cycles of Adriamycin and cyclophosphamide (4xAC) or four cycles of 4xAC and Paclitaxel (4xAC+4xPAC), aiming to unravel the mechanisms and cellular responses at these critical treatment stages. Notably, under normal and low glucose conditions, the resistant MDA-MB-231 cells showed accelerated growth compared to the control cells, while the resistant MCF7 cells proliferated more slowly than their original counterparts. Resistance to 4xAC resulted in significant cell death in both cell lines, especially under low glucose conditions, in contrast to control or 4xAC+4xPAC-resistant cells. The similarity between the MCF7 4xAC+4xPAC resistant cells and the control might be due to the P-AKT expression pattern in response to glucose levels since the levels were constant in MCF7 4xAC in all glucose concentrations. Molecular analysis revealed specific protein accumulations explaining the heightened proliferation and invasion in resistant MDA-MB-231 cells and their ability to withstand low glucose levels compared to MCF7. In conclusion, increased drug involvement corresponds to increased cell resistance, and changes in glucose levels differentially impact resistant variant cells to different drugs. The findings can be translated clinically to explain patients' differential responses to preoperative chemotherapy cycles considering their breast cancer subtype and diabetic status.
Collapse
Affiliation(s)
- Sirin A. Adham
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Azza Al Kalbani
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Noura Al Zeheimi
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Muna Al Dalali
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Noor Al Kharusi
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Azeeza Siddiqi
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Aliya Al Maskari
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
46
|
Mohammadi Zonouz A, Taghavi S, Nekooei S, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. Synthesis of targeted doxorubicin-loaded gold nanorod -mesoporous manganese dioxide core-shell nanostructure for ferroptosis, chemo-photothermal therapy in vitro and in vivo. Int J Pharm 2024; 665:124725. [PMID: 39293581 DOI: 10.1016/j.ijpharm.2024.124725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/14/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
In the current study, a core-shell inorganic nanostructure comprising a gold nanorod core and -mesoporous manganese dioxide shell was synthesized. Then, the mesoporous manganese dioxide shell was loaded with doxorubicin (DOX) and then coated with pluronic F127 and pluronic F127-folic acid conjugate (1.5:1 wt ratio of pluronic F127: pluronic F127-folic acid conjugate) to prepare targeted final platform. In this design, mesoporous manganese dioxide acted as a reservoir for DOX loading, anti-hypoxia, and MRI contrast agent, while the gold nanorod core acted as a photothermal and CT scan imaging agent. DOX was encapsulated in the mesoporous manganese dioxide shell with a loading capacity and loading efficiency of 19.8 % ± 0.2 and 99.0 % ± 0.9, respectively. The in vitro release experiment showed the impact of glutathione (GSH), mildly acidic pH, and laser irradiating toward accelerated stimuli-responsive DOX release. The ·OH production of the prepared platform was verified by methylene blue (MB) decomposition reaction. Furthermore, thermal imaging exhibited the ability of the prepared platform to convert the NIR irradiation to heat. In vitro cytotoxicity tests on the folate receptor-positive 4 T1 cell line revealed the remarkable cytotoxicity of the targeted formulation compared to the nontargeted formulation (statistically significant). The MTT experiment demonstrated that exposure to laser 808 irradiation enhanced cytotoxicity of the targeted formulation (p < 0.0001). The production of ROS in 4 T1 cells following treatment with the targeted formulation was demonstrated by the dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay. Furthermore, in vivo investigations by implementing subcutaneous 4 T1 tumorized female BABL/c mice indicated that the prepared platform was an effective system in suppressing tumor growth by combining chemotherapy with PTT (photothermal therapy). Additionally, simultanous PTT and anti-hypoxic activity of this system showed potent tumor growth suppression impact. The percent of tumor size reduction in mice treated with FA-F127-DOX@Au-MnO2 + 808 nm laser compared to the control group was 99.7 %. The results of the biodistribution investigation showed tumor accumulation and modified pharmacokinetics of the targeted system. Lastly, 6 and 24 h post-intravenous injection, CT-scan and MR imagings capability of the prepared platform was verified in preclinical stage. The prepared multipurpose system introduces great opportunity to provide multiple treatment strategy along with multimodal imaging capability in a single platform for breast cancer treatment.
Collapse
Affiliation(s)
- Aidin Mohammadi Zonouz
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Taghavi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medicinal Chemistry Department, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
47
|
Feng S, Zhang Y, Wang Y, Gao Y, Song Y. Harnessing Gene Editing Technology for Tumor Microenvironment Modulation: An Emerging Anticancer Strategy. Chemistry 2024; 30:e202402485. [PMID: 39225329 DOI: 10.1002/chem.202402485] [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: 06/29/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/04/2024]
Abstract
Cancer is a multifaceted disease influenced by both intrinsic cellular traits and extrinsic factors, with the tumor microenvironment (TME) being crucial for cancer progression. To satisfy their high proliferation and aggressiveness, cancer cells always plunder large amounts of nutrients and release various signals to their surroundings, forming a dynamic TME with special metabolic, immune, microbial and physical characteristics. Due to the neglect of interactions between tumor cells and the TME, traditional cancer therapies often struggle with challenges such as drug resistance, low efficacy, and recurrence. Importantly, the development of gene editing technologies, particularly the CRISPR-Cas system, offers promising new strategies for cancer treatment. Combined with nanomaterial strategies, CRISPR-Cas technology exhibits precision, affordability, and user-friendliness with reduced side effects, which holds great promise for profoundly altering the TME at the genetic level, potentially leading to lasting anticancer outcomes. This review will delve into how CRISPR-Cas can be leveraged to manipulate the TME, examining its potential as a transformative anticancer therapy.
Collapse
Affiliation(s)
- Shujun Feng
- College of Engineering and Applied Sciences, Nanjing University, 210023, Nanjing, China
| | - Yu Zhang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 210094, Nanjing, China
| | - Yanyi Wang
- College of Engineering and Applied Sciences, Nanjing University, 210023, Nanjing, China
| | - Yanfeng Gao
- School of Medical Imaging, Wannan Medical College, 241002, Wuhu, China
| | - Yujun Song
- College of Engineering and Applied Sciences, Nanjing University, 210023, Nanjing, China
| |
Collapse
|
48
|
Zhou Z, Dong D, Yuan Y, Luo J, Liu XD, Chen LY, Wang G, Yin Y. Single cell atlas reveals multilayered metabolic heterogeneity across tumour types. EBioMedicine 2024; 109:105389. [PMID: 39393173 DOI: 10.1016/j.ebiom.2024.105389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Metabolic reprogramming plays a pivotal role in cancer progression, contributing to substantial intratumour heterogeneity and influencing tumour behaviour. However, a systematic characterization of metabolic heterogeneity across multiple cancer types at the single-cell level remains limited. METHODS We integrated 296 tumour and normal samples spanning six common cancer types to construct a single-cell compendium of metabolic gene expression profiles and identify cell type-specific metabolic properties and reprogramming patterns. A computational approach based on non-negative matrix factorization (NMF) was utilised to identify metabolic meta-programs (MMPs) showing intratumour heterogeneity. In-vitro cell experiments were conducted to confirm the associations between MMPs and chemotherapy resistance, as well as the function of key metabolic regulators. Survival analyses were performed to assess clinical relevance of cellular metabolic properties. FINDINGS Our analysis revealed shared glycolysis upregulation and divergent regulation of citric acid cycle across different cell types. In malignant cells, we identified a colorectal cancer-specific MMP associated with resistance to the cuproptosis inducer elesclomol, validated through in-vitro cell experiments. Furthermore, our findings enabled the stratification of patients into distinct prognostic subtypes based on metabolic properties of specific cell types, such as myeloid cells. INTERPRETATION This study presents a nuanced understanding of multilayered metabolic heterogeneity, offering valuable insights into potential personalized therapies targeting tumour metabolism. FUNDING National Key Research and Development Program of China (2021YFA1300601). National Natural Science Foundation of China (key grants 82030081 and 81874235). The Shenzhen High-level Hospital Construction Fund and Shenzhen Basic Research Key Project (JCYJ20220818102811024). The Lam Chung Nin Foundation for Systems Biomedicine.
Collapse
Affiliation(s)
- Zhe Zhou
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Di Dong
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Yuyao Yuan
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Juan Luo
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Xiao-Ding Liu
- Research Centre for Molecular Pathology, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China
| | - Long-Yun Chen
- Research Centre for Molecular Pathology, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China
| | - Guangxi Wang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
49
|
Knudsen-Clark AM, Mwangi D, Cazarin J, Morris K, Baker C, Hablitz LM, McCall MN, Kim M, Altman BJ. Circadian rhythms of macrophages are altered by the acidic tumor microenvironment. EMBO Rep 2024; 25:5080-5112. [PMID: 39415049 PMCID: PMC11549407 DOI: 10.1038/s44319-024-00288-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are prime therapeutic targets due to their pro-tumorigenic functions, but varying efficacy of macrophage-targeting therapies highlights our incomplete understanding of how macrophages are regulated within the tumor microenvironment (TME). The circadian clock is a key regulator of macrophage function, but how circadian rhythms of macrophages are influenced by the TME remains unknown. Here, we show that conditions associated with the TME such as polarizing stimuli, acidic pH, and lactate can alter circadian rhythms in macrophages. While cyclic AMP (cAMP) has been reported to play a role in macrophage response to acidic pH, our results indicate pH-driven changes in circadian rhythms are not mediated solely by cAMP signaling. Remarkably, circadian disorder of TAMs was revealed by clock correlation distance analysis. Our data suggest that heterogeneity in circadian rhythms within the TAM population level may underlie this circadian disorder. Finally, we report that circadian regulation of macrophages suppresses tumor growth in a murine model of pancreatic cancer. Our work demonstrates a novel mechanism by which the TME influences macrophage biology through modulation of circadian rhythms.
Collapse
Affiliation(s)
- Amelia M Knudsen-Clark
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Daniel Mwangi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Juliana Cazarin
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Kristina Morris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Cameron Baker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Lauren M Hablitz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Matthew N McCall
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Brian J Altman
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
50
|
Khaliq AM, Rajamohan M, Saeed O, Mansouri K, Adil A, Zhang C, Turk A, Carstens JL, House M, Hayat S, Nagaraju GP, Pappas SG, Wang YA, Zyromski NJ, Opyrchal M, Lee KP, O'Hagan H, El Rayes B, Masood A. Spatial transcriptomic analysis of primary and metastatic pancreatic cancers highlights tumor microenvironmental heterogeneity. Nat Genet 2024; 56:2455-2465. [PMID: 39294496 DOI: 10.1038/s41588-024-01914-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/19/2024] [Indexed: 09/20/2024]
Abstract
Although the spatial, cellular and molecular landscapes of resected pancreatic ductal adenocarcinoma (PDAC) are well documented, the characteristics of its metastatic ecology remain elusive. By applying spatially resolved transcriptomics to matched primary and metastatic PDAC samples, we discovered a conserved continuum of fibrotic, metabolic and immunosuppressive spatial ecotypes across anatomical regions. We observed spatial tumor microenvironment heterogeneity spanning beyond that previously appreciated in PDAC. Through comparative analysis, we show that the spatial ecotypes exhibit distinct enrichment between primary and metastatic sites, implying adaptability to the local environment for survival and progression. The invasive border ecotype exhibits both pro-tumorigenic and anti-tumorigenic cell-type enrichment, suggesting a potential immunotherapy target. The ecotype heterogeneity across patients emphasizes the need to map individual patient landscapes to develop personalized treatment strategies. Collectively, our findings provide critical insights into metastatic PDAC biology and serve as a valuable resource for future therapeutic exploration and molecular investigations.
Collapse
Affiliation(s)
- Ateeq M Khaliq
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Meenakshi Rajamohan
- Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, IN, USA
| | - Omer Saeed
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kimia Mansouri
- Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, IN, USA
| | - Asif Adil
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chi Zhang
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anita Turk
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Julienne L Carstens
- Division of Hematology and Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael House
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Ganji P Nagaraju
- Division of Hematology and Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sam G Pappas
- Division of Surgical Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Y Alan Wang
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nicholas J Zyromski
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mateusz Opyrchal
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kelvin P Lee
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Heather O'Hagan
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bassel El Rayes
- Division of Hematology and Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ashiq Masood
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|