1
|
Abate-Shen C, Politi K. The Evolution of Mouse Models of Cancer: Past, Present, and Future. Cold Spring Harb Perspect Med 2025; 15:a041736. [PMID: 38772706 PMCID: PMC12047742 DOI: 10.1101/cshperspect.a041736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
In the nearly 50 years since the original models of cancer first hit the stage, mouse models have become a major contributor to virtually all aspects of cancer research, and these have evolved well beyond simple transgenic or xenograft models to encompass a wide range of more complex models. As the sophistication of mouse models has increased, an explosion of new technologies has expanded the potential to both further develop and apply these models to address major challenges in cancer research. In the current era, cancer modeling has expanded to include nongermline genetically engineered mouse models (GEMMs), patient-derived models, organoids, and adaptations of the models better suited for cancer immunology research. New technologies that have transformed the field include the application of CRISPR-Cas9-mediated genome editing, in vivo imaging, and single-cell analysis to cancer modeling. Here, we provide a historical perspective on the evolution of mouse models of cancer, focusing on how far we have come in a relatively short time and how new technologies will shape the future development of mouse models of cancer.
Collapse
Affiliation(s)
- Cory Abate-Shen
- Departments of Molecular Pharmacology and Therapeutics, Urology, Pathology and Cell Biology, Medicine, and Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - Katerina Politi
- Departments of Pathology and Internal Medicine (Section of Medical Oncology) and Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut 06405, USA
| |
Collapse
|
2
|
Oñate MK, Oon C, Bhattacharyya S, Low V, Chen C, Zhao X, Arnold F, Yan Z, Pramod S, Hang Y, Ho YJ, Lowe SW, Kim SK, Xia Z, Sherman MH. Stromal KITL/SCF Maintains Pancreas Tissue Homeostasis and Restrains Tumor Progression. Cancer Discov 2025; 15:913-929. [PMID: 39918337 PMCID: PMC12046321 DOI: 10.1158/2159-8290.cd-24-1079] [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: 07/29/2024] [Revised: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 02/12/2025]
Abstract
SIGNIFICANCE By analyzing transcriptional programs in healthy and tumor-associated pancreatic mesenchyme, we find that a subpopulation of mesenchymal cells in healthy pancreas tissue expresses the paracrine signaling factor KITL. The loss of mesenchymal KITL is an accompanying and permissive feature of pancreas tumor evolution, with potential implications for cancer interception. See related article by Dolskii and Cukierman, p. 872.
Collapse
Affiliation(s)
- Maria Kathrina Oñate
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Chet Oon
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Sohinee Bhattacharyya
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Vivien Low
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Canping Chen
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Xiaofan Zhao
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Frank Arnold
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Ziqiao Yan
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
| | - Sneha Pramod
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, New York, New York
| | - Yan Hang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California
| | - Yu-Jui Ho
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott W. Lowe
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Zheng Xia
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Mara H. Sherman
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
3
|
Zheng L, Cai W, Ke Y, Hu X, Yang C, Zhang R, Wu H, Liu D, Yu H, Wu C. Cancer‑associated fibroblasts: a pivotal regulator of tumor microenvironment in the context of radiotherapy. Cell Commun Signal 2025; 23:147. [PMID: 40114180 PMCID: PMC11927177 DOI: 10.1186/s12964-025-02138-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND In the course of tumor treatment, radiation therapy (RT) not only kills cancer cells, but also induces complex biological effects in non-malignant cells around cancer cells. These biological effects such as angiogenesis, changes in stromal composition and immune cell infiltration remodel the tumor microenvironment (TME). As one of the major components of the TME, Cancer‑associated fibroblasts (CAFs) are not only involved in tumorigenesis, progression, recurrence, and metastasis but also regulate the tumor-associated immune microenvironment. CAFs and tumor cells or immune cells have complex intercellular communication in the context of tumor radiation. MAIN CONTENT Different cellular precursors, spatial location differences, absence of specific markers, and advances in single-cell sequencing technology have gradually made the abundant heterogeneity of CAFs well known. Due to unique radioresistance properties, CAFs can survive under high doses of ionizing radiation. However, radiation can induce phenotypic and functional changes in CAFs and further act on tumor cells and immune cells to promote or inhibit tumor progression. To date, the effect of RT on CAFs and the effect of irradiated CAFs on tumor progression and TME are still not well defined. CONCLUSION In this review, we review the origin, phenotypic, and functional heterogeneity of CAFs and describe the effects of RT on CAFs, focusing on the mutual crosstalk between CAFs and tumor or immune cells after radiation. We also discuss emerging strategies for targeted CAFs therapy.
Collapse
Affiliation(s)
- Linhui Zheng
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Wenqi Cai
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Yuan Ke
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Xiaoyan Hu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Chunqian Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Runze Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Huachao Wu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Dong Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Haijun Yu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China.
| | - Chaoyan Wu
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
| |
Collapse
|
4
|
Kwon JY, Vera RE, Fernandez-Zapico ME. The multi-faceted roles of cancer-associated fibroblasts in pancreatic cancer. Cell Signal 2025; 127:111584. [PMID: 39756502 PMCID: PMC11807759 DOI: 10.1016/j.cellsig.2024.111584] [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] [Revised: 12/13/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
The tumor microenvironment (TME) has been linked with the pathogenesis of pancreatic ductal adenocarcinoma (PDAC), the most common histological subtype of pancreatic cancer. A central component of the TME are cancer-associated fibroblasts (CAFs), which can either suppress or promote tumor growth in a context-dependent manner. In this review, we will discuss the multi-faceted roles of CAFs in tumor-stroma interactions influencing cancer initiation, progression and therapeutic response.
Collapse
Affiliation(s)
- John Y Kwon
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN 55901, USA.
| | - Renzo E Vera
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN 55901, USA.
| | | |
Collapse
|
5
|
Nuñez-Rios JD, Ulrich H, Díaz-Muñoz M, Lameu C, Vázquez-Cuevas FG. Purinergic system in cancer stem cells. Purinergic Signal 2025; 21:23-38. [PMID: 37966629 PMCID: PMC11904000 DOI: 10.1007/s11302-023-09976-5] [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: 06/10/2023] [Accepted: 10/25/2023] [Indexed: 11/16/2023] Open
Abstract
Accumulating evidence supports the idea that cancer stem cells (CSCs) are those with the capacity to initiate tumors, generate phenotypical diversity, sustain growth, confer drug resistance, and orchestrate the spread of tumor cells. It is still controversial whether CSCs originate from normal stem cells residing in the tissue or cancer cells from the tumor bulk that have dedifferentiated to acquire stem-like characteristics. Although CSCs have been pointed out as key drivers in cancer, knowledge regarding their physiology is still blurry; thus, research focusing on CSCs is essential to designing novel and more effective therapeutics. The purinergic system has emerged as an important autocrine-paracrine messenger system with a prominent role at multiple levels of the tumor microenvironment, where it regulates cellular aspects of the tumors themselves and the stromal and immune systems. Recent findings have shown that purinergic signaling also participates in regulating the CSC phenotype. Here, we discuss updated information regarding CSCs in the purinergic system and present evidence supporting the idea that elements of the purinergic system expressed by this subpopulation of the tumor represent attractive pharmacological targets for proposing innovative anti-cancer therapies.
Collapse
Affiliation(s)
- J D Nuñez-Rios
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Juriquilla Querétaro, Querétaro, CP 76230, México
| | - H Ulrich
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - M Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Juriquilla Querétaro, Querétaro, CP 76230, México
| | - C Lameu
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - F G Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla #3001, Juriquilla Querétaro, Querétaro, CP 76230, México.
| |
Collapse
|
6
|
Yang Y, Sun H, Yu H, Wang L, Gao C, Mei H, Jiang X, Ji M. Tumor-associated-fibrosis and active collagen-CD44 axis characterize a poor-prognosis subtype of gastric cancer and contribute to tumor immunosuppression. J Transl Med 2025; 23:123. [PMID: 39871345 PMCID: PMC11773867 DOI: 10.1186/s12967-025-06070-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/04/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Tumor-associated fibrosis modifies the tumor microenvironment (TME), hinders the infiltration and activity of cytotoxic immune cells, and is a critical pathological process leading to the ineffectiveness of tumor immunotherapy in gastric cancer (GC). However, the specific mechanisms and interventions are yet to be fully explored. METHODS Our study included 375 gastric cancer samples from TCGA, 1 single-cell RNA sequencing (scRNA-seq) dataset comprising of 15 gastric cancer samples from GEO, 19 cohorts of immunotherapy and 2 GWAS datasets. Consensus clustering identified a gastric cancer subtype characterized primarily by fibrosis, and various methods such as pseudotime analysis, CellChat analysis and Colocalization analysis were used to explore its mechanisms. RESULTS A subtype of gastric cancer was identified with poor prognosis, characterized by higher malignancy, drug resistance, and poor immune infiltration, associated with elevated expression of genes related with Extracellular matrix (ECM). Single-cell transcriptome analysis showed active Collagen-CD44 signaling axis between cancer-associated fibroblasts (CAFs) and immune cells in gastric cancer, with ECM-related genes upregulated during tumor progression. The expression of CD44 was significantly elevated in the subtype, associated with poor prognosis and tumor immune suppression in gastric cancer, potentially involved in the recruitment of immunosuppressive cells such as M2 macrophages and regulatory T cells (Tregs) and the upregulation of multiple immune checkpoints including PD-1/PD-L1. CONCLUSION Our study identified a new subtype of gastric cancer, revealing that fibrosis is a critical mechanism driving immune suppression in gastric cancer and emphasizing the central role of the Collagen-CD44 signaling axis. The Collagen-CD44 signaling axis has the potential to serve as a novel therapeutic target for gastric cancer by enhancing immune cell-mediated tumor suppression. By combining it with immune checkpoint inhibitors (ICIs), it may improve the efficacy of immunotherapy for gastric cancer and offer new hope for treatment.
Collapse
Affiliation(s)
- Yingqi Yang
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Haohan Sun
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Hongkai Yu
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Luyao Wang
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Chang Gao
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Haokun Mei
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaomeng Jiang
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, China.
| | - Minghui Ji
- School of Nursing, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
7
|
Zhou S, Zhao Z, Wang Z, Xu H, Li Y, Xu K, Li W, Yang J. Cancer-associated fibroblasts in carcinogenesis. J Transl Med 2025; 23:50. [PMID: 39806363 PMCID: PMC11727299 DOI: 10.1186/s12967-025-06071-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
In contemporary times, cancer poses the most significant threat to human life and safety. Scientists have relentlessly pursued the intricacies of carcinogenesis and explored ways to prevent and treat cancer. Carcinogenesis is a complex, multi-faceted, and multi-stage process, with numerous underlying causes, including inflammation and fibrosis. Cancer-associated fibroblasts (CAFs), however, occupy a pivotal and substantial role within the tumor microenvironment, facilitating carcinogenesis through diverse mechanisms such as creating inflammation, fostering a fibrotic tumor microenvironment, and immunosuppression. In this paper, we introduce the concept of carcinogenesis, explain its causes, describe the characteristics of CAFs and their sources, and highlight the roles and mechanisms of CAFs in promoting carcinogenesis. Ultimately, our aim is to contribute to the development of novel therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Shufen Zhou
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zekun Zhao
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhaojun Wang
- Department of Thyroid and Breast Surgery, The DingLi Clinical, The Wenzhou Central Hospital, College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hanzheng Xu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yijie Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China.
| | - Wei Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Jiahua Yang
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|
8
|
Van Keymeulen A. Mechanisms of Regulation of Cell Fate in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:167-184. [PMID: 39821026 DOI: 10.1007/978-3-031-70875-6_10] [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: 01/19/2025]
Abstract
This chapter focuses on the mechanisms of regulation of cell fate in breast development, occurring mainly after birth, as well as in breast cancer. First, we will review how the microenvironment of the breast, as well as external cues, plays a crucial role in mammary gland cell specification and will describe how it has been shown to reprogram non-mammary cells into mammary epithelial cells. Then we will focus on the transcription factors and master regulators which have been established to be determinant for basal (BC) and luminal cell (LC) identity, and will describe the experiments of ectopic expression or loss of function of these transcription factors which demonstrated that they were crucial for cell fate. We will also discuss how master regulators are involved in the fate choice of LCs between estrogen receptor (ER)-positive cells and ER- cells, which will give rise to alveolar cells upon pregnancy and lactation. We will describe how oncogene expression induces reprogramming and change of fate of mammary epithelial cells before tumor appearance, which could be an essential step in tumorigenesis. Finally, we will describe the involvement of master regulators of mammary epithelial cells in breast cancer.
Collapse
Affiliation(s)
- Alexandra Van Keymeulen
- Laboratory of Stem Cells and Cancer (LSCC), Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
9
|
Ingthorsson S, Traustadottir GA, Gudjonsson T. Breast Morphogenesis: From Normal Development to Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:29-44. [PMID: 39821019 DOI: 10.1007/978-3-031-70875-6_3] [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: 01/19/2025]
Abstract
The human breast gland is composed of branching epithelial ducts that culminate in milk-producing units known as terminal duct lobular units (TDLUs). The epithelial compartment comprises an inner layer of luminal epithelial cells (LEP) and an outer layer of contractile myoepithelial cells (MEP). Both LEP and MEP arise from a common stem cell population. The epithelial compartment undergoes dynamic branching morphogenesis and remodelling, which expands the surface area for milk production. The epithelial remodelling that starts at the onset of menarche is largely under hormonal control, first and foremost by estrogen and progesterone from ovaries, the production of which is stimulated by pituitary-derived hormones. Menopause leads to a significant decline in estrogen and progesterone levels, resulting in involution and senescence of the breast epithelium. The branching morphogenesis involves developmental events such as epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET). EMT and MET confer plasticity to the epithelial compartment enabling the migration of epithelial cells through the stroma and restoration of the epithelial phenotype. In the normal breast, the stroma, including the basement membrane (BM), collagen-rich extracellular matrix, and various stromal cells, supports the correct histoarchitecture of the glandular tree. However, in cancer, the stroma can acquire tumour-promoting properties and is referred to as the tumour microenvironment. This chapter will explore the developmental processes including branching morphogenesis in the normal breast gland and discuss the lineage relationship between LEPS and MEPs and their interactions with the surrounding stroma in the normal and neoplastic breast gland. Finally, we will review various in vitro and in vivo models employed in mammary gland research.
Collapse
Affiliation(s)
- Saevar Ingthorsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Faculty of Nursing and Midwifery, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Gunnhildur Asta Traustadottir
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Pathology, Landspitali University Hospital, Reykjavik, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
- Department of Laboratory Hematology, Landspitali University Hospital, Reykjavik, Iceland.
| |
Collapse
|
10
|
Iggo R, MacGrogan G. Classification of Breast Cancer Through the Perspective of Cell Identity Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:185-207. [PMID: 39821027 DOI: 10.1007/978-3-031-70875-6_11] [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: 01/19/2025]
Abstract
The mammary epithelium has an inner luminal layer that contains estrogen receptor (ER)-positive hormone-sensing cells and ER-negative alveolar/secretory cells, and an outer basal layer that contains myoepithelial/stem cells. Most human tumours resemble either hormone-sensing cells or alveolar/secretory cells. The most widely used molecular classification, the Intrinsic classification, assigns hormone-sensing tumours to Luminal A/B and human epidermal growth factor 2-enriched (HER2E)/molecular apocrine (MA)/luminal androgen receptor (LAR)-positive classes, and alveolar/secretory tumours to the Basal-like class. Molecular classification is most useful when tumours have classic invasive carcinoma of no special type (NST) histology. It is less useful for special histological types of breast cancer, such as metaplastic breast cancer and adenoid cystic cancer, which are better described with standard pathology terms. Compared to mice, humans show a strong bias towards making tumours that resemble mammary hormone-sensing cells. This could be caused by the formation in adolescence of der(1;16), a translocation through the centromeres of chromosomes 1 and 16, which only occurs in humans and could trap the cells in the hormone-sensing state.
Collapse
Affiliation(s)
- Richard Iggo
- INSERM, Bergonie Cancer Institute, University of Bordeaux, Bordeaux, France.
| | - Gaetan MacGrogan
- INSERM, Bergonie Cancer Institute, University of Bordeaux, Bordeaux, France
| |
Collapse
|
11
|
Rinkenbaugh AL, Qi Y, Cai S, Shao J, Hancock F, Jeter-Jones S, Zhang X, Powell E, Huo L, Lau R, Fu C, Gould R, den Hollander P, Ravenberg EE, White JB, Rauch GM, Arun B, Yam C, Thompson AM, Echeverria GV, Moulder SL, Symmans WF, Chang JT, Piwnica-Worms H. An annotated biobank of triple negative breast cancer patient-derived xenografts featuring treatment-naïve and longitudinal samples throughout neoadjuvant chemotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625287. [PMID: 39651299 PMCID: PMC11623626 DOI: 10.1101/2024.11.25.625287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Triple negative breast cancer (TNBC) that fails to respond to neoadjuvant chemotherapy (NACT) can be lethal. Developing effective strategies to eradicate chemoresistant disease requires experimental models that recapitulate the heterogeneity characteristic of TNBC. To that end, we established a biobank of 92 orthotopic patient-derived xenograft (PDX) models of TNBC from the tumors of 75 patients enrolled in the ARTEMIS clinical trial ( NCT02276443 ) at MD Anderson Cancer Center, including 12 longitudinal sets generated from serial patient biopsies collected throughout NACT and from metastatic disease. Models were established from both chemosensitive and chemoresistant tumors, and nearly 30% of PDX models were capable of lung metastasis. Comprehensive molecular profiling demonstrated conservation of genomes and transcriptomes between patient and corresponding PDX tumors, with representation of all major transcriptional subtypes. Transcriptional changes observed in the longitudinal PDX models highlight dysregulation in pathways associated with DNA integrity, extracellular matrix interactions, the ubiquitin-proteasome system, epigenetics, and inflammatory signaling. These alterations reveal a complex network of adaptations associated with chemoresistance. This PDX biobank provides a valuable resource for tackling the most pressing issues facing the clinical management of TNBC, namely chemoresistance and metastasis.
Collapse
|
12
|
Graham S, Dmitrieva M, Vendramini-Costa DB, Francescone R, Trujillo MA, Cukierman E, Wood LD. From precursor to cancer: decoding the intrinsic and extrinsic pathways of pancreatic intraepithelial neoplasia progression. Carcinogenesis 2024; 45:801-816. [PMID: 39514554 PMCID: PMC12098012 DOI: 10.1093/carcin/bgae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/04/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
This review explores the progression of pancreatic intraepithelial neoplasia (PanIN) to pancreatic ductal adenocarcinoma through a dual lens of intrinsic molecular alterations and extrinsic microenvironmental influences. PanIN development begins with Kirsten rat sarcoma viral oncogene (KRAS) mutations driving PanIN initiation. Key additional mutations in cyclin-dependent kinase inhibitor 2A (CDKN2A), tumor protein p53 (TP53), and mothers against decapentaplegic homolog 4 (SMAD4) disrupt cell cycle control and genomic stability, crucial for PanIN progression from low-grade to high-grade dysplasia. Additional molecular alterations in neoplastic cells, including epigenetic modifications and chromosomal alterations, can further contribute to neoplastic progression. In parallel with these alterations in neoplastic cells, the microenvironment, including fibroblast activation, extracellular matrix remodeling, and immune modulation, plays a pivotal role in PanIN initiation and progression. Crosstalk between neoplastic and stromal cells influences nutrient support and immune evasion, contributing to tumor development, growth, and survival. This review underscores the intricate interplay between cell-intrinsic molecular drivers and cell-extrinsic microenvironmental factors, shaping PanIN predisposition, initiation, and progression. Future research aims to unravel these interactions to develop targeted therapeutic strategies and early detection techniques, aiming to alleviate the severe impact of pancreatic cancer by addressing both genetic predispositions and environmental influences.
Collapse
Affiliation(s)
- Sarah Graham
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Mariia Dmitrieva
- Cancer Signaling & Microenvironment Program, M&C Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA 19111, United States
| | - Debora Barbosa Vendramini-Costa
- Henry Ford Pancreatic Cancer Center, Henry Ford Health, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, United States
| | - Ralph Francescone
- Henry Ford Pancreatic Cancer Center, Henry Ford Health, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, United States
| | - Maria A Trujillo
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Edna Cukierman
- Cancer Signaling & Microenvironment Program, M&C Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA 19111, United States
| | - Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD 21231, United States
| |
Collapse
|
13
|
Finot L, Hue-Beauvais C, Aujean E, Le Provost F, Chanat E. Sorted stem/progenitor epithelial cells of pubertal bovine mammary gland present limited potential to reconstitute an organised mammary epithelium after transplantation. PLoS One 2024; 19:e0296614. [PMID: 39423190 PMCID: PMC11488748 DOI: 10.1371/journal.pone.0296614] [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: 12/20/2023] [Accepted: 09/29/2024] [Indexed: 10/21/2024] Open
Abstract
The development and maintenance of mammary gland tissue depend on the proliferation and differentiation of mammary stem and progenitor cells. Here, we investigated populations of mammary epithelial cells that are potential candidates for bovine mammary gland development using xenotransplantation into mice cleared mammary fat pad. Transplanted mammary explants from 17-month-old Holstein heifers developed outgrowths exhibiting the archetypal morphology and molecular marker distributions of the bovine gland. Xenotransplantation of sorted mammary epithelial cells (CD49fpos) into bovinised fat pads using inactivated bovine fibroblasts resulted in outgrowth developments with 50% take rate, but these lacked the ductal or alveolar epithelial structures of the normal mammary gland. Similar results were obtained with xenografts of candidate bovine mammary epithelial stem cells (CD49fhighCD24pos) or epithelial cells of the basal lineage (CD49fhighCD24neg) which also developed as clumps of cells surrounded by stromal stretches within the mouse adipose tissue. In conclusion, sorted cells showed compromised regenerative potential for epithelial morphogenesis. Further work is therefore needed to identify mammary stem/progenitor cells with full regenerative capabilities for biogenesis of normal mammary gland structure, with milk-secreting function.
Collapse
Affiliation(s)
- Laurence Finot
- PEGASE, INRAE, Institut Agro, Saint Gilles, Paris, France
| | - Cathy Hue-Beauvais
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, Paris, France
| | - Etienne Aujean
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, Paris, France
| | - Fabienne Le Provost
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, Paris, France
| | - Eric Chanat
- PEGASE, INRAE, Institut Agro, Saint Gilles, Paris, France
| |
Collapse
|
14
|
Zhang H, Li S, Wang D, Liu S, Xiao T, Gu W, Yang H, Wang H, Yang M, Chen P. Metabolic reprogramming and immune evasion: the interplay in the tumor microenvironment. Biomark Res 2024; 12:96. [PMID: 39227970 PMCID: PMC11373140 DOI: 10.1186/s40364-024-00646-1] [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: 07/29/2024] [Accepted: 08/24/2024] [Indexed: 09/05/2024] Open
Abstract
Tumor cells possess complex immune evasion mechanisms to evade immune system attacks, primarily through metabolic reprogramming, which significantly alters the tumor microenvironment (TME) to modulate immune cell functions. When a tumor is sufficiently immunogenic, it can activate cytotoxic T-cells to target and destroy it. However, tumors adapt by manipulating their metabolic pathways, particularly glucose, amino acid, and lipid metabolism, to create an immunosuppressive TME that promotes immune escape. These metabolic alterations impact the function and differentiation of non-tumor cells within the TME, such as inhibiting effector T-cell activity while expanding regulatory T-cells and myeloid-derived suppressor cells. Additionally, these changes lead to an imbalance in cytokine and chemokine secretion, further enhancing the immunosuppressive landscape. Emerging research is increasingly focusing on the regulatory roles of non-tumor cells within the TME, evaluating how their reprogrammed glucose, amino acid, and lipid metabolism influence their functional changes and ultimately aid in tumor immune evasion. Despite our incomplete understanding of the intricate metabolic interactions between tumor and non-tumor cells, the connection between these elements presents significant challenges for cancer immunotherapy. This review highlights the impact of altered glucose, amino acid, and lipid metabolism in the TME on the metabolism and function of non-tumor cells, providing new insights that could facilitate the development of novel cancer immunotherapies.
Collapse
Affiliation(s)
- Haixia Zhang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Shizhen Li
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Dan Wang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Siyang Liu
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Tengfei Xiao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Wangning Gu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Hongmin Yang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Hui Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China.
| | - Minghua Yang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China.
| | - Pan Chen
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China.
| |
Collapse
|
15
|
Valdivia-Silva J, Chinney-Herrera A. Chemokine receptors and their ligands in breast cancer: The key roles in progression and metastasis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 388:124-161. [PMID: 39260935 DOI: 10.1016/bs.ircmb.2024.07.002] [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: 09/13/2024]
Abstract
Chemokines and their receptors are a family of chemotactic cytokines with important functions in the immune response in both health and disease. Their known physiological roles such as the regulation of leukocyte trafficking and the development of immune organs generated great interest when it was found that they were also related to the control of early and late inflammatory stages in the tumor microenvironment. In fact, in breast cancer, an imbalance in the synthesis of chemokines and/or in the expression of their receptors was attributed to be involved in the regulation of disease progression, including invasion and metastasis. Research in this area is progressing rapidly and the development of new agents based on chemokine and chemokine receptor antagonists are emerging as attractive alternative strategies. This chapter provides a snapshot of the different functions reported for chemokines and their receptors with respect to the potential to regulate breast cancer progression.
Collapse
Affiliation(s)
- Julio Valdivia-Silva
- Centro de Investigación en Bioingenieria (BIO), Universidad de Ingenieria y Tecnologia-UTEC, Barranco, Lima, Peru.
| | - Alberto Chinney-Herrera
- Facultad de Medicina, Universidad Nacional Autonoma de Mexico-UNAM, Ciudad Universitaria, Coyoacan, Ciudad de Mexico, Mexico
| |
Collapse
|
16
|
Kaufmann J, Haist M, Kur IM, Zimmer S, Hagemann J, Matthias C, Grabbe S, Schmidberger H, Weigert A, Mayer A. Tumor-stroma contact ratio - a novel predictive factor for tumor response to chemoradiotherapy in locally advanced oropharyngeal cancer. Transl Oncol 2024; 46:102019. [PMID: 38833784 PMCID: PMC11190748 DOI: 10.1016/j.tranon.2024.102019] [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: 04/19/2023] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/06/2024] Open
Abstract
The growth pattern of oropharyngeal squamous cell carcinomas (OPSCC) varies from compact tumor cell aggregates to diffusely infiltrating tumor cell-clusters. The influence of the growth pattern on local tumor control and survival has been studied mainly for surgically treated oral cavity carcinomas on a visual basis. In this study, we used multiplex immunofluorescence staining (mIF) to examine the antigens pan-cytokeratin, p16INK4a, Ki67, CD271, PD-L1, and CD8 in pretherapeutic biopsies from 86 OPSCC. We introduce Tumor-stroma contact ratio (TSC), a novel parameter, to quantify the relationship between tumor cells in contact with the stromal surface and the total number of epithelial tumor cells. mIF tumor cores were analyzed at the single-cell level, and tumor-stromal contact area was quantified using the R package "Spatstat". TSC was correlated with the visually assessed invasion pattern by two independent investigators. Furthermore, TSC was analyzed in relation to clinical parameters and patient survival data to evaluate its potential prognostic significance. Higher TSC correlated with poor response to (chemo-)radiotherapy (r = 0.3, p < 0.01), and shorter overall (OS) and progression-free (PFS) survival (median OS: 13 vs 136 months, p < 0.0001; median PFS: 5 vs 85 months, p < 0.0001). Visual categorization of growth pattern according to established criteria of tumor aggressiveness showed interobserver variability increasing with more nuanced categories (2 categories: k = 0.7, 95 %-CI: 0.55 - 0.85; 4 categories k = 0.48, 95 %-CI: 0.35 - 0.61). In conclusion, TSC is an objective and reproducible computer-based parameter to quantify tumor-stroma contact area. We demonstrate its relevance for the response of oropharyngeal carcinomas to primary (chemo-)radiotherapy.
Collapse
Affiliation(s)
- Justus Kaufmann
- Department of Radiation Oncology and Radiotherapy, University Medical Center of the Johannes-Gutenberg-University, Mainz 55131, Germany.
| | - Maximilian Haist
- Department of Dermatology, University Medical Center of the Johannes-Gutenberg-University, 55131 Mainz, Germany; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ivan-Maximiliano Kur
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60596 Frankfurt, Germany
| | - Stefanie Zimmer
- Institute of Pathology, University Medical Center of the Johannes-Gutenberg-University, 55131 Mainz, Germany
| | - Jan Hagemann
- Department of Otorhinolaryngology, University Medical Center of the Johannes-Gutenberg-University, Mainz 55131, Germany
| | - Christoph Matthias
- Department of Otorhinolaryngology, University Medical Center of the Johannes-Gutenberg-University, Mainz 55131, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes-Gutenberg-University, 55131 Mainz, Germany
| | - Heinz Schmidberger
- Department of Radiation Oncology and Radiotherapy, University Medical Center of the Johannes-Gutenberg-University, Mainz 55131, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60596 Frankfurt, Germany
| | - Arnulf Mayer
- Department of Radiation Oncology and Radiotherapy, University Medical Center of the Johannes-Gutenberg-University, Mainz 55131, Germany; Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
17
|
Oñate MK, Oon C, Bhattacharyya S, Low V, Chen C, Zhao X, Yan Z, Hang Y, Kim SK, Xia Z, Sherman MH. Stromal KITL/SCF promotes pancreas tissue homeostasis and restrains tumor progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605485. [PMID: 39131374 PMCID: PMC11312444 DOI: 10.1101/2024.07.29.605485] [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: 08/13/2024]
Abstract
Components of normal tissue architecture serve as barriers to tumor progression. Inflammatory and wound-healing programs are requisite features of solid tumorigenesis, wherein alterations to immune and non-immune stromal elements enable loss of homeostasis during tumor evolution. The precise mechanisms by which normal stromal cell states limit tissue plasticity and tumorigenesis, and which are lost during tumor progression, remain largely unknown. Here we show that healthy pancreatic mesenchyme expresses the paracrine signaling molecule KITL, also known as stem cell factor, and identify loss of stromal KITL during tumorigenesis as tumor-promoting. Genetic inhibition of mesenchymal KITL in the contexts of homeostasis, injury, and cancer together indicate a role for KITL signaling in maintenance of pancreas tissue architecture, such that loss of the stromal KITL pool increased tumor growth and reduced survival of tumor-bearing mice. Together, these findings implicate loss of mesenchymal KITL as a mechanism for establishing a tumor-permissive microenvironment.
Collapse
Affiliation(s)
- M. Kathrina Oñate
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Chet Oon
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Sohinee Bhattacharyya
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Vivien Low
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Canping Chen
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Xiaofan Zhao
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Ziqiao Yan
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
| | - Yan Hang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Zheng Xia
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Mara H. Sherman
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
18
|
Khan IR, Sadida HQ, Hashem S, Singh M, Macha MA, Al-Shabeeb Akil AS, Khurshid I, Bhat AA. Therapeutic implications of signaling pathways and tumor microenvironment interactions in esophageal cancer. Biomed Pharmacother 2024; 176:116873. [PMID: 38843587 DOI: 10.1016/j.biopha.2024.116873] [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: 03/24/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Esophageal cancer (EC) is significantly influenced by the tumor microenvironment (TME) and altered signaling pathways. Downregulating these pathways in EC is essential for suppressing tumor development, preventing metastasis, and enhancing therapeutic outcomes. This approach can increase tumor sensitivity to treatments, enhance patient outcomes, and inhibit cancer cell proliferation and spread. The TME, comprising cellular and non-cellular elements surrounding the tumor, significantly influences EC's development, course, and treatment responsiveness. Understanding the complex relationships within the TME is crucial for developing successful EC treatments. Immunotherapy is a vital TME treatment for EC. However, the heterogeneity within the TME limits the application of anticancer drugs outside clinical settings. Therefore, identifying reliable microenvironmental biomarkers that can detect therapeutic responses before initiating therapy is crucial. Combining approaches focusing on EC signaling pathways with TME can enhance treatment outcomes. This integrated strategy aims to interfere with essential signaling pathways promoting cancer spread while disrupting factors encouraging tumor development. Unraveling aberrant signaling pathways and TME components can lead to more focused and efficient treatment approaches, identifying specific cellular targets for treatments. Targeting the TME and signaling pathways may reduce metastasis risk by interfering with mechanisms facilitating cancer cell invasion and dissemination. In conclusion, this integrative strategy has significant potential for improving patient outcomes and advancing EC research and therapy. This review discusses the altered signaling pathways and TME in EC, focusing on potential future therapeutics.
Collapse
Affiliation(s)
- Inamu Rashid Khan
- Department of Zoology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine Doha 26999, Qatar
| | - Mayank Singh
- Department of Medical Oncology (Lab), Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu and Kashmir 192122, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar
| | - Ibraq Khurshid
- Department of Zoology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar.
| |
Collapse
|
19
|
Vera MJ, Ponce I, Almarza C, Ramirez G, Guajardo F, Dubois-Camacho K, Tobar N, Urra FA, Martinez J. CCL2 and Lactate from Chemotherapeutics-Treated Fibroblasts Drive Malignant Traits by Metabolic Rewiring in Low-Migrating Breast Cancer Cell Lines. Antioxidants (Basel) 2024; 13:801. [PMID: 39061870 PMCID: PMC11274190 DOI: 10.3390/antiox13070801] [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: 05/07/2024] [Revised: 06/10/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
While cytostatic chemotherapy targeting DNA is known to induce genotoxicity, leading to cell cycle arrest and cytokine secretion, the impact of these drugs on fibroblast-epithelial cancer cell communication and metabolism remains understudied. Our research focused on human breast fibroblast RMF-621 exposed to nonlethal concentrations of cisplatin and doxorubicin, revealing reduced proliferation, diminished basal and maximal mitochondrial respirations, heightened mitochondrial ROS and lactate production, and elevated MCT4 protein levels. Interestingly, RMF-621 cells enhanced glucose uptake, promoting lactate export. Breast cancer cells MCF-7 exposed to conditioned media (CM) from drug-treated stromal RMF-621 cells increased MCT1 protein levels, lactate-driven mitochondrial respiration, and a significantly high mitochondrial spare capacity for lactate. These changes occurred alongside altered mitochondrial respiration, mitochondrial membrane potential, and superoxide levels. Furthermore, CM with doxorubicin and cisplatin increased migratory capacity in MCF-7 cells, which was inhibited by MCT1 (BAY-8002), glutamate dehydrogenase (EGCG), mitochondrial pyruvate carrier (UK5099), and complex I (rotenone) inhibitors. A similar behavior was observed in T47-D and ZR-75-1 breast cancer cells. This suggests that CM induces metabolic rewiring involving elevated lactate uptake to sustain mitochondrial bioenergetics during migration. Treatment with the mitochondrial-targeting antioxidant mitoTEMPO in RMF-621 and the addition of an anti-CCL2 antibody in the CM prevented the promigratory MCF-7 phenotype. Similar effects were observed in THP1 monocyte cells, where CM increased monocyte recruitment. We propose that nonlethal concentrations of DNA-damaging drugs induce changes in the cellular environment favoring a promalignant state dependent on mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Maria Jesus Vera
- Laboratory of Cellular Biology, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
- Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca 3480094, Chile
| | - Iván Ponce
- Laboratory of Cellular Biology, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
| | - Cristopher Almarza
- Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca 3480094, Chile
- Laboratory of Metabolic Plasticity and Bioenergetics, Program of Molecular and Clinical Pharmacology, Institute of Biomedical Science (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago 7810000, Chile
- Network for Snake Venom Research and Drug Discovery, Santiago 7810000, Chile
| | - Gonzalo Ramirez
- Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca 3480094, Chile
- Laboratory of Metabolic Plasticity and Bioenergetics, Program of Molecular and Clinical Pharmacology, Institute of Biomedical Science (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago 7810000, Chile
- Network for Snake Venom Research and Drug Discovery, Santiago 7810000, Chile
| | - Francisco Guajardo
- Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca 3480094, Chile
- Laboratory of Metabolic Plasticity and Bioenergetics, Program of Molecular and Clinical Pharmacology, Institute of Biomedical Science (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago 7810000, Chile
- Network for Snake Venom Research and Drug Discovery, Santiago 7810000, Chile
| | - Karen Dubois-Camacho
- Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca 3480094, Chile
- Laboratory of Metabolic Plasticity and Bioenergetics, Program of Molecular and Clinical Pharmacology, Institute of Biomedical Science (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago 7810000, Chile
- Network for Snake Venom Research and Drug Discovery, Santiago 7810000, Chile
| | - Nicolás Tobar
- Laboratory of Cellular Biology, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
| | - Félix A. Urra
- Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca 3480094, Chile
- Laboratory of Metabolic Plasticity and Bioenergetics, Program of Molecular and Clinical Pharmacology, Institute of Biomedical Science (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago 7810000, Chile
- Network for Snake Venom Research and Drug Discovery, Santiago 7810000, Chile
- Interuniversity Center for Healthy Aging (CIES), Consortium of Universities of the State of Chile (CUECH), Santiago 8320216, Chile
| | - Jorge Martinez
- Laboratory of Cellular Biology, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
- Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca 3480094, Chile
| |
Collapse
|
20
|
Bu W, Li Y. Advances in Immunocompetent Mouse and Rat Models. Cold Spring Harb Perspect Med 2024; 14:a041328. [PMID: 37217281 PMCID: PMC10810718 DOI: 10.1101/cshperspect.a041328] [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] [Indexed: 05/24/2023]
Abstract
Rodent models of breast cancer have played critical roles in our understanding of breast cancer development and progression as well as preclinical testing of cancer prevention and therapeutics. In this article, we first review the values and challenges of conventional genetically engineered mouse (GEM) models and newer iterations of these models, especially those with inducible or conditional regulation of oncogenes and tumor suppressors. Then, we discuss nongermline (somatic) GEM models of breast cancer with temporospatial control, made possible by intraductal injection of viral vectors to deliver oncogenes or to manipulate the genome of mammary epithelial cells. Next, we introduce the latest development in precision editing of endogenous genes using in vivo CRISPR-Cas9 technology. We conclude with the recent development in generating somatic rat models for modeling estrogen receptor-positive breast cancer, something that has been difficult to accomplish in mice.
Collapse
Affiliation(s)
- Wen Bu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yi Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
21
|
Miller JL, Reddy A, Harman RM, Van de Walle GR. A xenotransplantation mouse model to study physiology of the mammary gland from large mammals. PLoS One 2024; 19:e0298390. [PMID: 38416747 PMCID: PMC10901318 DOI: 10.1371/journal.pone.0298390] [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: 09/06/2023] [Accepted: 01/23/2024] [Indexed: 03/01/2024] Open
Abstract
Although highly conserved in structure and function, many (patho)physiological processes of the mammary gland vary drastically between mammals, with mechanisms regulating these differences not well understood. Large mammals display variable lactation strategies and mammary cancer incidence, however, research into these variations is often limited to in vitro analysis due to logistical limitations. Validating a model with functional mammary xenografts from cryopreserved tissue fragments would allow for in vivo comparative analysis of mammary glands from large and/or rare mammals and would improve our understanding of postnatal development, lactation, and premalignancy across mammals. To this end, we generated functional mammary xenografts using mammary tissue fragments containing mammary stroma and parenchyma isolated via an antibody-independent approach from healthy, nulliparous equine and canine donor tissues to study these species in vivo. Cryopreserved mammary tissue fragments were xenotransplanted into de-epithelialized fat pads of immunodeficient mice and resulting xenografts were structurally and functionally assessed. Preimplantation of mammary stromal fibroblasts was performed to promote ductal morphogenesis. Xenografts recapitulated mammary lobule architecture and contained donor-derived stromal components. Mammatropic hormone stimulation resulted in (i) upregulation of lactation-associated genes, (ii) altered proliferation index, and (iii) morphological changes, indicating functionality. Preimplantation of mammary stromal fibroblasts did not promote ductal morphogenesis. This model presents the opportunity to study novel mechanisms regulating unique lactation strategies and mammary cancer induction in vivo. Due to the universal applicability of this approach, this model serves as proof-of-concept for developing mammary xenografts for in vivo analysis of virtually any mammals, including large and rare mammals.
Collapse
Affiliation(s)
- James L Miller
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Alexandra Reddy
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
22
|
Ortiz JR, Lewis SM, Ciccone M, Chatterjee D, Henry S, Siepel A, Dos Santos CO. Single-Cell Transcription Mapping of Murine and Human Mammary Organoids Responses to Female Hormones. J Mammary Gland Biol Neoplasia 2024; 29:3. [PMID: 38289401 PMCID: PMC10827859 DOI: 10.1007/s10911-023-09553-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/18/2023] [Indexed: 02/01/2024] Open
Abstract
During female adolescence and pregnancy, rising levels of hormones result in a cyclic source of signals that control the development of mammary tissue. While such alterations are well understood from a whole-gland perspective, the alterations that such hormones bring to organoid cultures derived from mammary glands have yet to be fully mapped. This is of special importance given that organoids are considered suitable systems to understand cross species breast development. Here we utilized single-cell transcriptional profiling to delineate responses of murine and human normal breast organoid systems to female hormones across evolutionary distinct species. Collectively, our study represents a molecular atlas of epithelial dynamics in response to estrogen and pregnancy hormones.
Collapse
Affiliation(s)
| | - Steven M Lewis
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Michael Ciccone
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | | | - Samantha Henry
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Adam Siepel
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | | |
Collapse
|
23
|
Sitte Z, Miranda Buzetta AA, Jones SJ, Lin ZW, Whitman NA, Lockett MR. Paper-Based Coculture Platform to Evaluate the Effects of Fibroblasts on Estrogen Signaling in ER+ Breast Cancers. ACS MEASUREMENT SCIENCE AU 2023; 3:479-487. [PMID: 38145029 PMCID: PMC10740124 DOI: 10.1021/acsmeasuresciau.3c00032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 12/26/2023]
Abstract
Cell-based assays enable molecular-level studies of cellular responses to drug candidates or potential toxins. Transactivation assays quantify the activation or inhibition of nuclear receptors, key transcriptional regulators of gene targets in mamalian cells. One such assay couples the expression of luciferase to the transcriptional activity of estrogen receptor-alpha (ERα). While this assay is regularly used to screen for agonists and antagonists of the estrogen signaling pathway, the setup relies on monolayer cultures in which cells are plated directly onto the surface of cell-compatible plasticware. The tumor microenvironment is more than a collection of cancerous cells and is profoundly influenced by tissue architecture, the presence of extracellular matrices, and intercellular signaling molecules produced by non-cancerous neighboring cells (e.g., fibroblasts). There exists a need for three-dimensional culture platforms that can be rapidly prototyped to assess new configurations and readily produced in the large numbers needed for translational studies and screening applications. Here, we demonstrate the utility of the paper-based culture platform to probe the effects of intercellular signaling between two cell types. We used paper scaffolds to generate tumor-like environments, forming a defined volume of breast cancer cells suspended in collagen. By placing the paper scaffolds in commercial 96-well plates, we compared monocultures of only breast cancer cells with coculture configurations containing fibroblasts in different locations that mimicked the stages of breast cancer progression. We show that ERα transactivation in the T47D-KBluc cell line is affected by the presence, number, and proximity of fibroblasts, and is a consequence of intercellular signaling molecules. After screening a small library of fibroblast-secreted signaling molecules, we showed that interleukin-6 (IL-6) was the primary driver of reduced estradiol sensitivity. These effects were mitigated in the coculture configurations by the addition of an IL-6 neutralizing antibody. We also assessed estrogen receptor expression and transcriptional regulation, further demonstrating the utility of the paper-based platform for detailed mechanistic studies.
Collapse
Affiliation(s)
- Zachary
R. Sitte
- Department
of Chemistry, University of North Carolina
at Chapel Hill, Kenan and Caudill Laboratories, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
| | - Abel Andre Miranda Buzetta
- Department
of Chemistry, University of North Carolina
at Chapel Hill, Kenan and Caudill Laboratories, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
| | - Sarina J. Jones
- Department
of Chemistry, University of North Carolina
at Chapel Hill, Kenan and Caudill Laboratories, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
| | - Zhi-Wei Lin
- Department
of Chemistry, University of North Carolina
at Chapel Hill, Kenan and Caudill Laboratories, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
| | - Nathan Ashbrook Whitman
- Department
of Chemistry, University of North Carolina
at Chapel Hill, Kenan and Caudill Laboratories, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
| | - Matthew R. Lockett
- Department
of Chemistry, University of North Carolina
at Chapel Hill, Kenan and Caudill Laboratories, 125 South Road, Chapel Hill, North Carolina 27599-3290, United States
- Lineberger
Comprehensive Cancer Center, University
of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill, North Carolina 27599-7295, United States
| |
Collapse
|
24
|
Gao D, Fang L, Liu C, Yang M, Yu X, Wang L, Zhang W, Sun C, Zhuang J. Microenvironmental regulation in tumor progression: Interactions between cancer-associated fibroblasts and immune cells. Biomed Pharmacother 2023; 167:115622. [PMID: 37783155 DOI: 10.1016/j.biopha.2023.115622] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/20/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023] Open
Abstract
The tumor microenvironment (TME), the "soil" on which tumor cells grow, has an important role in regulating the proliferation and metastasis of tumor cells as well as their response to treatment. Cancer-associated fibroblasts (CAFs), as the most abundant stromal cells of the TME, can not only directly alter the immunosuppressive effect of the TME through their own metabolism, but also influence the aggregation and function of immune cells by secreting a large number of cytokines and chemokines, reducing the body's immune surveillance of tumor cells and making them more prone to immune escape. Our study provides a comprehensive review of fibroblast chemotaxis, malignant transformation, metabolic characteristics, and interactions with immune cells. In addition, the current small molecule drugs targeting CAFs have been summarized, including both natural small molecules and targeted drugs for current clinical therapeutic applications. A complete review of the role of fibroblasts in TME from an immune perspective is presented, which has important implications in improving the efficiency of immunotherapy by targeting fibroblasts.
Collapse
Affiliation(s)
- Dandan Gao
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Liguang Fang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Mengrui Yang
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Xiaoyun Yu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Longyun Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Wenfeng Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China; Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| |
Collapse
|
25
|
Lin S, Margueron R, Charafe-Jauffret E, Ginestier C. Disruption of lineage integrity as a precursor to breast tumor initiation. Trends Cell Biol 2023; 33:887-897. [PMID: 37061355 DOI: 10.1016/j.tcb.2023.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 04/17/2023]
Abstract
Increase in lineage infidelity and/or imbalance is frequently observed around the earliest stage of breast tumor initiation. In response to disruption of homeostasis, differentiated cells can partially lose their identity and gain cellular plasticity, a process involving epigenome landscape remodeling. This increase of cellular plasticity may promote the malignant transformation of breast tumors and fuel their heterogeneity. Here, we review recent studies that have yield insights into important regulators of lineage integrity and mechanisms that trigger mammary epithelial lineage derail, and evaluate their impacts on breast tumor development.
Collapse
Affiliation(s)
- Shuheng Lin
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille Univeristy, Epithelial Stem Cells and Cancer Laboratory, Equipe Labellisée LIGUE Contre le Cancer, Marseille, France
| | - Raphaël Margueron
- Institut Curie, PSL Research University, Sorbonne University, Paris, France
| | - Emmanuelle Charafe-Jauffret
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille Univeristy, Epithelial Stem Cells and Cancer Laboratory, Equipe Labellisée LIGUE Contre le Cancer, Marseille, France.
| | - Christophe Ginestier
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille Univeristy, Epithelial Stem Cells and Cancer Laboratory, Equipe Labellisée LIGUE Contre le Cancer, Marseille, France.
| |
Collapse
|
26
|
Ortiz JR, Lewis SM, Ciccone MF, Chatterjee D, Henry S, Siepel A, Dos Santos CO. Single-cell transcription mapping of murine and human mammary organoids responses to female hormones. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.559971. [PMID: 37808747 PMCID: PMC10557705 DOI: 10.1101/2023.09.28.559971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
During female adolescence and pregnancy, rising levels of hormones result in a cyclic source of signals that control the development of mammary tissue. While such alterations are well understood from a whole-gland perspective, the alterations that such hormones bring to organoid cultures derived from mammary glands have yet to be fully mapped. This is of special importance given that organoids are considered suitable systems to understand cross species breast development. Here we utilized single-cell transcriptional profiling to delineate responses of murine and human normal breast organoid systems to female hormones across evolutionary distinct species. Collectively, our study represents a molecular atlas of epithelial dynamics in response to estrogen and pregnancy hormones.
Collapse
|
27
|
Zhao Y, Shen M, Wu L, Yang H, Yao Y, Yang Q, Du J, Liu L, Li Y, Bai Y. Stromal cells in the tumor microenvironment: accomplices of tumor progression? Cell Death Dis 2023; 14:587. [PMID: 37666813 PMCID: PMC10477351 DOI: 10.1038/s41419-023-06110-6] [Citation(s) in RCA: 136] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023]
Abstract
The tumor microenvironment (TME) is made up of cells and extracellular matrix (non-cellular component), and cellular components include cancer cells and non-malignant cells such as immune cells and stromal cells. These three types of cells establish complex signals in the body and further influence tumor genesis, development, metastasis and participate in resistance to anti-tumor therapy. It has attracted scholars to study immune cells in TME due to the significant efficacy of immune checkpoint inhibitors (ICI) and chimeric antigen receptor T (CAR-T) in solid tumors and hematologic tumors. After more than 10 years of efforts, the role of immune cells in TME and the strategy of treating tumors based on immune cells have developed rapidly. Moreover, ICI have been recommended by guidelines as first- or second-line treatment strategies in a variety of tumors. At the same time, stromal cells is another major class of cellular components in TME, which also play a very important role in tumor metabolism, growth, metastasis, immune evasion and treatment resistance. Stromal cells can be recruited from neighboring non-cancerous host stromal cells and can also be formed by transdifferentiation from stromal cells to stromal cells or from tumor cells to stromal cells. Moreover, they participate in tumor genesis, development and drug resistance by secreting various factors and exosomes, participating in tumor angiogenesis and tumor metabolism, regulating the immune response in TME and extracellular matrix. However, with the deepening understanding of stromal cells, people found that stromal cells not only have the effect of promoting tumor but also can inhibit tumor in some cases. In this review, we will introduce the origin of stromal cells in TME as well as the role and specific mechanism of stromal cells in tumorigenesis and tumor development and strategies for treatment of tumors based on stromal cells. We will focus on tumor-associated fibroblasts (CAFs), mesenchymal stem cells (MSCs), tumor-associated adipocytes (CAAs), tumor endothelial cells (TECs) and pericytes (PCs) in stromal cells.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, 130033, Changchun, Jilin, China
| | - Meili Shen
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, 130033, Changchun, Jilin, China
| | - Liangqiang Wu
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, 130012, Changchun, Jilin, China
| | - Haiqin Yang
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, 130012, Changchun, Jilin, China
| | - Yixuan Yao
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, 130012, Changchun, Jilin, China
| | - Qingbiao Yang
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, 130012, Changchun, Jilin, China
| | - Jianshi Du
- Key Laboratory of Lymphatic Surgery Jilin Province, Jilin Engineering Laboratory for Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin University, 130033, Changchun, Jilin, China
| | - Linlin Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, 130033, Changchun, Jilin, China
| | - Yapeng Li
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, 130012, Changchun, Jilin, China.
| | - Yuansong Bai
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, 130033, Changchun, Jilin, China.
| |
Collapse
|
28
|
Zhang W, Tao N, Bai L. Polysaccharides from Lentinus edodes prevent acquired drug resistance to docetaxel in prostate cancer cells by decreasing the TGF-β1 secretion of cancer-associated fibroblasts. J Nat Med 2023; 77:817-828. [PMID: 37354258 DOI: 10.1007/s11418-023-01722-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/11/2023] [Indexed: 06/26/2023]
Abstract
Prostate cancer is one of the most prevalent lethal diseases among men globally. In the treatment of prostate cancer, the limited therapeutic efficacy of the standard non-hormonal systemic therapy docetaxel (DTX) represents an important challenge. Cancer-associated fibroblasts (CAFs) play a crucial role in resistance to therapy because of their prevalence and functional pleiotropy in tumor environments. Our previous research revealed that MPSSS, a novel polysaccharide extracted from Lentinus edodes, could significantly attenuate the immunosuppressive function of myeloid suppressor cells and CAFs. In this study, we investigated whether MPSSS could potentiate the efficacy of DTX against prostate cancer by inhibiting CAF-induced chemoresistance and elucidated its underlying mechanisms. The sensitivity of PC-3 prostate cancer cells cultured with conditioned medium derived from CAFs (CAF-CM) to DTX was assessed. The resistance effect induced by CAF-CM was abolished when CAFs were pretreated with MPSSS. Bioinformatic analysis of datasets from the Gene Expression Omnibus database revealed the activation of the transforming growth factor β1 (TGF-β1) signaling pathway in DTX-resistant cells. Based on this finding, we demonstrated that treatment with the TGF-β1 receptor inhibitor SB525334 reversed DTX resistance in CAFs, suggesting that TGF-β1 secreted by CAFs was a crucial intermediary in the development of DTX resistance in PC3 cells. Further research revealed that MPSSS decreases the secretion of TGF-β1 by inhibiting the JAK2/STAT3 pathway via Toll-like receptor 4 in CAFs. Overall, MPSSS might be a potential adjuvant treatment for DTX resistance in prostate cancer.
Collapse
Affiliation(s)
- Wensheng Zhang
- Chinese PLA medical school, Beijing, China
- Department of Oncology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Ning Tao
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| | - Li Bai
- Chinese PLA medical school, Beijing, China.
- Department of Oncology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
29
|
Koch MK, Ravichandran A, Murekatete B, Clegg J, Joseph MT, Hampson M, Jenkinson M, Bauer HS, Snell C, Liu C, Gough M, Thompson EW, Werner C, Hutmacher DW, Haupt LM, Bray LJ. Exploring the Potential of PEG-Heparin Hydrogels to Support Long-Term Ex Vivo Culture of Patient-Derived Breast Explant Tissues. Adv Healthc Mater 2023; 12:e2202202. [PMID: 36527735 PMCID: PMC11469079 DOI: 10.1002/adhm.202202202] [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: 08/29/2022] [Revised: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Breast cancer is a complex, highly heterogenous, and dynamic disease and the leading cause of cancer-related death in women worldwide. Evaluation of the heterogeneity of breast cancer and its various subtypes is crucial to identify novel treatment strategies that can overcome the limitations of currently available options. Explant cultures of human mammary tissue have been known to provide important insights for the study of breast cancer structure and phenotype as they include the context of the surrounding microenvironment, allowing for the comprehensive exploration of patient heterogeneity. However, the major limitation of currently available techniques remains the short-term viability of the tissue owing to loss of structural integrity. Here, an ex vivo culture model using star-shaped poly(ethylene glycol) and maleimide-functionalized heparin (PEG-HM) hydrogels to provide structural support to the explant cultures is presented. The mechanical support allows the culture of the human mammary tissue for up to 3 weeks and prevent disintegration of the cellular structures including the epithelium and surrounding stromal tissue. Further, maintenance of epithelial phenotype and hormonal receptors is observed for up to 2 weeks of culture which makes them relevant for testing therapeutic interventions. Through this study, the importance of donor-to-donor variability and intra-patient tissue heterogeneity is reiterated.
Collapse
Affiliation(s)
- Maria K. Koch
- School of MechanicalMedical and Process EngineeringQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
| | - Akhilandeshwari Ravichandran
- School of MechanicalMedical and Process EngineeringQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
- Centre for Biomedical TechnologiesQueensland University of Technology (QUT)BrisbaneQLD4059Australia
| | - Berline Murekatete
- School of MechanicalMedical and Process EngineeringQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
| | - Julien Clegg
- School of MechanicalMedical and Process EngineeringQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
- Centre for the Personalised Analysis of CancersQueensland University of Technology (QUT)Translational Research InstituteBrisbaneQLD4102Australia
| | - Mary Teresa Joseph
- School of MechanicalMedical and Process EngineeringQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
| | - Madison Hampson
- School of MechanicalMedical and Process EngineeringQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
| | - Mitchell Jenkinson
- School of MechanicalMedical and Process EngineeringQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
| | - Hannah S. Bauer
- School of MechanicalMedical and Process EngineeringQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
| | - Cameron Snell
- Peter MacCallum Cancer CentreMelbourneVIC3000Australia
- Mater PathologyMater Hospital BrisbaneMater Health ServicesBrisbaneQLD4101Australia
| | - Cheng Liu
- Mater PathologyMater Hospital BrisbaneMater Health ServicesBrisbaneQLD4101Australia
- Faculty of MedicineThe University of QueenslandHerstonQLD4006Australia
| | - Madeline Gough
- Mater PathologyMater Hospital BrisbaneMater Health ServicesBrisbaneQLD4101Australia
- Cancer Pathology Research GroupMater Research Institute – The University of QueenslandTranslational Research InstituteBrisbaneQLD4102Australia
| | - Erik W. Thompson
- Centre for the Personalised Analysis of CancersQueensland University of Technology (QUT)Translational Research InstituteBrisbaneQLD4102Australia
- School of Biomedical SciencesQueensland University of Technology (QUT)Translational Research InstituteBrisbaneQLD4102Australia
| | - Carsten Werner
- Leibniz Institute of Polymer Research01069DresdenGermany
| | - Dietmar W. Hutmacher
- School of MechanicalMedical and Process EngineeringQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering TechnologiesQueensland University of Technology (QUT)BrisbaneQLD4000Australia
- Australian Research Council (ARC) Training Centre for Multiscale 3D ImagingModellingand Manufacturing (M3D Innovation)Queensland University of Technology (QUT)BrisbaneQLD4000Australia
- Max Planck Queensland Center for the Materials Science of Extracellular MatricesQueensland University of Technology (QUT)BrisbaneQLD4000Australia
| | - Larisa M. Haupt
- School of Biomedical SciencesQueensland University of Technology (QUT)Translational Research InstituteBrisbaneQLD4102Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering TechnologiesQueensland University of Technology (QUT)BrisbaneQLD4000Australia
- Max Planck Queensland Center for the Materials Science of Extracellular MatricesQueensland University of Technology (QUT)BrisbaneQLD4000Australia
- Centre for Genomics and Personalised HealthGenomics Research CentreSchool of Biomedical SciencesQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
| | - Laura J. Bray
- School of MechanicalMedical and Process EngineeringQueensland University of Technology (QUT)Kelvin GroveQLD4059Australia
- Centre for Biomedical TechnologiesQueensland University of Technology (QUT)BrisbaneQLD4059Australia
- Centre for the Personalised Analysis of CancersQueensland University of Technology (QUT)Translational Research InstituteBrisbaneQLD4102Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering TechnologiesQueensland University of Technology (QUT)BrisbaneQLD4000Australia
- Max Planck Queensland Center for the Materials Science of Extracellular MatricesQueensland University of Technology (QUT)BrisbaneQLD4000Australia
| |
Collapse
|
30
|
Verstappe J, Berx G. A role for partial epithelial-to-mesenchymal transition in enabling stemness in homeostasis and cancer. Semin Cancer Biol 2023; 90:15-28. [PMID: 36773819 DOI: 10.1016/j.semcancer.2023.02.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/19/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023]
Abstract
Stem cells have self-renewal capacities and the ability to give rise to differentiated cells thereby sustaining tissues during homeostasis and injury. This structural hierarchy extends to tumours which harbor stem-like cells deemed cancer stem cells that propagate the tumour and drive metastasis and relapse. The process of epithelial-to-mesenchymal transition (EMT), which plays an important role in development and cancer cell migration, was shown to be correlated with stemness in both homeostasis and cancer indicating that stemness can be acquired and is not necessarily an intrinsic trait. Nowadays it is experimentally proven that the activation of an EMT program does not necessarily drive cells towards a fully mesenchymal phenotype but rather to hybrid E/M states. This review offers the latest advances in connecting the EMT status and stem-cell state of both non-transformed and cancer cells. Recent literature clearly shows that hybrid EMT states have a higher probability of acquiring stem cell traits. The position of a cell along the EMT-axis which coincides with a stem cell-like state is known as the stemness window. We show how the original EMT-state of a cell dictates the EMT/MET inducing programmes required to reach stemness. Lastly we present the mechanism of stemness regulation and the regulatory feedback loops which position cells at a certain EMT state along the EMT axis.
Collapse
Affiliation(s)
- Jeroen Verstappe
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Geert Berx
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
31
|
Monaco ML, Idris OA, Essani K. Triple-Negative Breast Cancer: Basic Biology and Immuno-Oncolytic Viruses. Cancers (Basel) 2023; 15:cancers15082393. [PMID: 37190321 DOI: 10.3390/cancers15082393] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most lethal subtype of breast cancer. TNBC diagnoses account for approximately one-fifth of all breast cancer cases globally. The lack of receptors for estrogen, progesterone, and human epidermal growth factor 2 (HER-2, CD340) results in a lack of available molecular-based therapeutics. This increases the difficulty of treatment and leaves more traditional as well as toxic therapies as the only available standards of care in many cases. Recurrence is an additional serious problem, contributing substantially to its higher mortality rate as compared to other breast cancers. Tumor heterogeneity also poses a large obstacle to treatment approaches. No driver of tumor development has been identified for TNBC, and large variations in mutational burden between tumors have been described previously. Here, we describe the biology of six different subtypes of TNBC, based on differential gene expression. Subtype differences can have a large impact on metastatic potential and resistance to treatment. Emerging antibody-based therapeutics, such as immune checkpoint inhibitors, have available targets for small subsets of TNBC patients, leading to partial responses and relatively low overall efficacy. Immuno-oncolytic viruses (OVs) have recently become significant in the pursuit of effective treatments for TNBC. OVs generally share the ability to ignore the heterogeneous nature of TNBC cells and allow infection throughout a treated tumor. Recent genetic engineering has allowed for the enhancement of efficacy against certain tumor types while avoiding the most common side effects in non-cancerous tissues. In this review, TNBC is described in order to address the challenges it presents to potential treatments. The OVs currently described preclinically and in various stages of clinical trials are also summarized, as are their strategies to enhance therapeutic potential.
Collapse
Affiliation(s)
- Michael L Monaco
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| | - Omer A Idris
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| |
Collapse
|
32
|
Cao C, Lu X, Guo X, Zhao H, Gao Y. Patient-derived models: Promising tools for accelerating the clinical translation of breast cancer research findings. Exp Cell Res 2023; 425:113538. [PMID: 36871856 DOI: 10.1016/j.yexcr.2023.113538] [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: 12/12/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023]
Abstract
Breast cancer has become the highest incidence of cancer in women. It was extensively and deeply studied by biologists and medical workers worldwide. However, the meaningful results in lab researches cannot be realized in clinical, and a part of new drugs in clinical experiments do not obtain as good results as the preclinical researches. It is urgently that promote a kind of breast cancer research models that can get study results closer to the physiological condition of the human body. Patient-derived models (PDMs) originating from clinical tumor, contain primary elements of tumor and maintain key clinical features of tumor. So they are promising research models to facilitate laboratory researches translate to clinical application, and predict the treatment outcome of patients. In this review, we summarize the establishment of PDMs of breast cancer, reviewed the application of PDMs in clinical translational researches and personalized precision medicine with breast cancer as an example, to improve the understanding of PDMs among researchers and clinician, facilitate them to use PDMs on a large scale of breast cancer researches and promote the clinical translation of laboratory research and new drug development.
Collapse
Affiliation(s)
- Changqing Cao
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, China; State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, China
| | - Xiyan Lu
- Department of Outpatient, The Second Affiliated Hospital of Air Force Medical University, China
| | - Xinyan Guo
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, China
| | - Huadong Zhao
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, China.
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, China.
| |
Collapse
|
33
|
Kang J, Su M, Xu Q, Wang C, Yuan X, Han Z. Tumour-stroma ratio is a valuable prognostic factor for oral tongue squamous cell carcinoma. Oral Dis 2023; 29:628-638. [PMID: 34455659 DOI: 10.1111/odi.14013] [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: 06/04/2021] [Revised: 08/08/2021] [Accepted: 08/17/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The objectives of this study were to estimate the prognostic value of the tumour-stroma ratio (TSR) and tumour budding (TB) in oral tongue squamous cell carcinoma (OTSCC) and to establish a reliable model to predict the outcome of OTSCC patients. METHODS A total of 103 patients surgically treated at our hospital were enrolled in this study. Chi-square tests, Kaplan-Meier analyses and Cox proportional hazards regression models were performed for statistical analysis. RESULTS Fifty-six patients were categorized as stroma-rich, and 47 patients were categorized as stroma-poor. Only pathological grade was associated with the TSR (p = 0.017). Kaplan-Meier analysis showed that stroma-rich, high-intensity budding and high risk groups were associated with worse prognosis. The Cox regression model showed that the TSR was an independent risk factor for OTSCC patients prognosis, and the high risk group was also related to poor prognosis (p < 0.05). TB was significantly associated with poor prognosis but was not an independent risk factor. CONCLUSIONS We found that patients in the stroma-rich group had a worse long-term prognosis. The TSR is an independent risk factor for OTSCC patients' outcome. In addition, a risk model that combined the TSR and TB proved to be valuable for predicting OTSCC patients' outcome.
Collapse
Affiliation(s)
- Jia Kang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Ming Su
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Qiaoshi Xu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Chong Wang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Xiaohong Yuan
- Department of Pathology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Zhengxue Han
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
34
|
Pei L, Liu Y, Liu L, Gao S, Gao X, Feng Y, Sun Z, Zhang Y, Wang C. Roles of cancer-associated fibroblasts (CAFs) in anti- PD-1/PD-L1 immunotherapy for solid cancers. Mol Cancer 2023; 22:29. [PMID: 36759842 PMCID: PMC9912573 DOI: 10.1186/s12943-023-01731-z] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
In recent years, breakthroughs have been made in tumor immunotherapy. However, tumor immunotherapy, particularly anti-PD-1/PD-L1 immune checkpoint inhibitors, is effective in only a small percentage of patients in solid cancer. How to improve the efficiency of cancer immunotherapy is an urgent problem to be solved. As we all know, the state of the tumor microenvironment (TME) is an essential factor affecting the effectiveness of tumor immunotherapy, and the cancer-associated fibroblasts (CAFs) in TME have attracted much attention in recent years. As one of the main components of TME, CAFs interact with cancer cells and immune cells by secreting cytokines and vesicles, participating in ECM remodeling, and finally affecting the immune response process. With the in-depth study of CAFs heterogeneity, new strategies are provided for finding targets of combination immunotherapy and predicting immune efficacy. In this review, we focus on the role of CAFs in the solid cancer immune microenvironment, and then further elaborate on the potential mechanisms and pathways of CAFs influencing anti-PD-1/PD-L1 immunotherapy. In addition, we summarize the potential clinical application value of CAFs-related targets and markers in solid cancers.
Collapse
Affiliation(s)
- Liping Pei
- grid.412633.10000 0004 1799 0733Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China ,grid.412633.10000 0004 1799 0733Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yang Liu
- grid.414008.90000 0004 1799 4638Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Lin Liu
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Shuochen Gao
- grid.412633.10000 0004 1799 0733Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Xueyan Gao
- grid.412633.10000 0004 1799 0733Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yudi Feng
- grid.412633.10000 0004 1799 0733Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Zhenqiang Sun
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yan Zhang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Chengzeng Wang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
35
|
Echeverria GV, Cai S, Tu Y, Shao J, Powell E, Redwood AB, Jiang Y, McCoy A, Rinkenbaugh AL, Lau R, Trevarton AJ, Fu C, Gould R, Ravenberg EE, Huo L, Candelaria R, Santiago L, Adrada BE, Lane DL, Rauch GM, Yang WT, White JB, Chang JT, Moulder SL, Symmans WF, Hilsenbeck SG, Piwnica-Worms H. Predictors of success in establishing orthotopic patient-derived xenograft models of triple negative breast cancer. NPJ Breast Cancer 2023; 9:2. [PMID: 36627285 PMCID: PMC9831981 DOI: 10.1038/s41523-022-00502-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Patient-derived xenograft (PDX) models of breast cancer are an effective discovery platform and tool for preclinical pharmacologic testing and biomarker identification. We established orthotopic PDX models of triple negative breast cancer (TNBC) from the primary breast tumors of patients prior to and following neoadjuvant chemotherapy (NACT) while they were enrolled in the ARTEMIS trial (NCT02276443). Serial biopsies were obtained from patients prior to treatment (pre-NACT), from poorly responsive disease after four cycles of Adriamycin and cyclophosphamide (AC, mid-NACT), and in cases of AC-resistance, after a 3-month course of different experimental therapies and/or additional chemotherapy (post-NACT). Our study cohort includes a total of 269 fine needle aspirates (FNAs) from 217 women, generating a total of 62 PDX models (overall success-rate = 23%). Success of PDX engraftment was generally higher from those cancers that proved to be treatment-resistant, whether poorly responsive to AC as determined by ultrasound measurements mid-NACT (p = 0.063), RCB II/III status after NACT (p = 0.046), or metastatic relapse within 2 years of surgery (p = 0.008). TNBC molecular subtype determined from gene expression microarrays of pre-NACT tumors revealed no significant association with PDX engraftment rate (p = 0.877). Finally, we developed a statistical model predictive of PDX engraftment using percent Ki67 positive cells in the patient's diagnostic biopsy, positive lymph node status at diagnosis, and low volumetric reduction of the patient's tumor following AC treatment. This novel bank of 62 PDX models of TNBC provides a valuable resource for biomarker discovery and preclinical therapeutic trials aimed at improving neoadjuvant response rates for patients with TNBC.
Collapse
Affiliation(s)
- Gloria V Echeverria
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Lester and Sue Smith Breast Cancer Center and Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Shirong Cai
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yizheng Tu
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jiansu Shao
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Emily Powell
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Abena B Redwood
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yan Jiang
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Aaron McCoy
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Amanda L Rinkenbaugh
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rosanna Lau
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Alexander J Trevarton
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chunxiao Fu
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rebekah Gould
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Elizabeth E Ravenberg
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Lei Huo
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rosalind Candelaria
- Department of Breast Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Lumarie Santiago
- Department of Breast Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Beatriz E Adrada
- Department of Breast Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Deanna L Lane
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gaiane M Rauch
- Department of Abdominal Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wei T Yang
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jason B White
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Stacy L Moulder
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - W Fraser Symmans
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
36
|
Muacevic A, Adler JR, Ghenev P. Fibroblast Growth Factor-2 and the Invasive Potential in Urothelial Malignancies of the Bladder. Cureus 2023; 15:e34147. [PMID: 36843751 PMCID: PMC9949347 DOI: 10.7759/cureus.34147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction Urothelial carcinomas represent a distinct group of malignancies with a high recurrence potential. Multiple studies have established a set of interactions between the tumor cells of urothelial neoplasms and the extracellular matrix regarding invasion and tumor progression. In the present study, we evaluated the expression of fibroblast growth factor-2 (FGF2) in early-stage urothelial carcinomas of the urinary bladder (pTa and pT1) regarding the invasive potential of these tumors. Materials and methods A retrospective non-clinical approach was utilized for the study. Tumor tissue sections used for the initial diagnosis were stained by immunohistochemical means with an anti-FGF2 antibody and the expression within the extracellular matrix was evaluated using a histo-score (h-score). Statistical parameters regarding tumor invasion, FGF2 expression pattern and levels, patient demographic characteristics, and disease recurrence were analyzed for significance. Results A total of 163 cases were analyzed, with an h-score of 110 determined as the optimal cut-off value for invasive potential regarding FGF2 expression, with a sensitivity of 75.4% and a specificity of 78.9%. No statistical correlation was established between the demographic profile of the patients and the occurrence of disease recurrence. Conclusion Based on our results, the study of tumor-extracellular matrix interactions in regards to FGF2 expression is a promising field, at least in urothelial malignancies of the urinary bladder, in regards to tumor invasive potential, while it remains unestablished how these interactions affect metastatic potential.
Collapse
|
37
|
Azimian Zavareh V, Rafiee L, Sheikholeslam M, Shariati L, Vaseghi G, Savoji H, Haghjooy Javanmard S. Three-Dimensional in Vitro Models: A Promising Tool To Scale-Up Breast Cancer Research. ACS Biomater Sci Eng 2022; 8:4648-4672. [PMID: 36260561 DOI: 10.1021/acsbiomaterials.2c00277] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Common models used in breast cancer studies, including two-dimensional (2D) cultures and animal models, do not precisely model all aspects of breast tumors. These models do not well simulate the cell-cell and cell-stromal interactions required for normal tumor growth in the body and lake tumor like microenvironment. Three-dimensional (3D) cell culture models are novel approaches to studying breast cancer. They do not have the restrictions of these conventional models and are able to recapitulate the structural architecture, complexity, and specific function of breast tumors and provide similar in vivo responses to therapeutic regimens. These models can be a link between former traditional 2D culture and in vivo models and are necessary for further studies in cancer. This review attempts to summarize the most common 3D in vitro models used in breast cancer studies, including scaffold-free (spheroid and organoid), scaffold-based, and chip-based models, particularly focused on the basic and translational application of these 3D models in drug screening and the tumor microenvironment in breast cancer.
Collapse
Affiliation(s)
- Vajihe Azimian Zavareh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran.,Core Research Facilities (CRF), Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Laleh Rafiee
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Mohammadali Sheikholeslam
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran.,Biosensor Research Center, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran.,Cancer Prevention Research Center, Omid Hospital, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Golnaz Vaseghi
- Isfahan Cardiovascular Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Houman Savoji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada.,Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC H3T 1C5, Canada.,Montreal TransMedTech Institute, Montreal, QC H3T 1J4, Canada
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| |
Collapse
|
38
|
Mir S, Ormsbee Golden BD, Griess BJ, Vengoji R, Tom E, Kosmacek EA, Oberley-Deegan RE, Talmon GA, Band V, Teoh-Fitzgerald ML. Upregulation of Nox4 induces a pro-survival Nrf2 response in cancer-associated fibroblasts that promotes tumorigenesis and metastasis, in part via Birc5 induction. Breast Cancer Res 2022; 24:48. [PMID: 35836253 PMCID: PMC9281082 DOI: 10.1186/s13058-022-01548-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/30/2022] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND A pro-oxidant enzyme, NADPH oxidase 4 (Nox4) has been reported to be a critical downstream effector of TGFβ-induced myofibroblast transformation during fibrosis. While there are a small number of studies suggesting an oncogenic role of Nox4 derived from activated fibroblasts, direct evidence linking this pro-oxidant to the tumor-supporting CAF phenotype and the mechanisms involved are lacking, particularly in breast cancer. METHODS We targeted Nox4 in breast patient-derived CAFs via siRNA-mediated knockdown or administration of a pharmaceutical inhibitor (GKT137831). We also determine primary tumor growth and metastasis of implanted tumor cells using a stable Nox4-/- syngeneic mouse model. Autophagic flux of CAFs was assessed using a tandem fluorescent-tagged ptfl-LC3 plasmid via confocal microscopy analysis and determination of the expression level of autophagy markers (beclin-1 and LC3B). Nox4 overexpressing CAFs depend on the Nrf2 (nuclear factor-erythroid factor 2-related factor 2) pathway for survival. We then determined the dependency of Nox4-overexpressing CAFs on the Nrf2-mediated adaptive stress response pathway for survival. Furthermore, we investigated the involvement of Birc5 on CAF phenotype (viability and collagen contraction activity) as well as the expression level of CAF markers, FAP and αSMA. CONCLUSIONS We found that deletion of stroma Nox4 and pharmaceutically targeting its activity with GKT137831 significantly inhibited orthotopic tumor growth and metastasis of implanted E0771 and 4T1 murine mammary carcinoma cell lines in mice. More importantly, we found a significant upregulation of Nox4 expression in CAFs isolated from human breast tumors versus normal mammary fibroblasts (RMFs). Our in situ RNA hybridization analysis for Nox4 transcription on a human breast tumor microarray further support a role of this pro-oxidant in the stroma of breast carcinomas. In addition, we found that Nox4 promotes autophagy in CAFs. Moreover, we found that Nox4 promoted survival of CAFs via activation of Nrf2, a master regulator of oxidative stress response. We have further shown Birc5 is involved as a downstream modulator of Nrf2-mediated pro-survival phenotype. Together these studies indicate a role of redox signaling via the Nox4-Nrf2 pathway in tumorigenesis and metastasis of breast cancer cells by promoting autophagy and survival of CAFs.
Collapse
Affiliation(s)
- Shakeel Mir
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Briana D Ormsbee Golden
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Brandon J Griess
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Eric Tom
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Vimla Band
- Department of Genetics, Cell Biology and Anatomy, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Melissa Lt Teoh-Fitzgerald
- Department of Biochemistry and Molecular Biology, Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, 7005 Durham Research Center, 985870 Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
39
|
Current Nano-Strategies to Target Tumor Microenvironment to Improve Anti-Tumor efficiency. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
40
|
Cadamuro M, Fabris L, Zhang X, Strazzabosco M. Tumor microenvironment and immunology of cholangiocarcinoma. HEPATOMA RESEARCH 2022; 8:11. [PMID: 39301518 PMCID: PMC11412615 DOI: 10.20517/2394-5079.2021.140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Cholangiocarcinoma (CCA), an aggressive tumor originating from both intra- and extra-hepatic biliary cells, represents an unmet need in liver oncology, as treatment remains largely unsatisfactory. A typical feature of CCA is the presence of a complex tumor microenvironment (TME) composed of neoplastic cells, a rich inflammatory infiltrate, and cancer-associated fibroblasts and desmoplastic matrix that makes it extremely chemoresistant to traditional chemotherapeutic drugs. In this review, we describe the cell populations within the TME, in particular those involved in the innate and adaptive immune response and how they interact with tumor cells and with matrix proteins. The TME is crucial for CCA to mount an immune escape response and is the battlefield where molecularly targeted therapies and immune therapy, particularly in combination, may actually prove their therapeutic value.
Collapse
Affiliation(s)
| | - Luca Fabris
- Department of Molecular Medicine (DMM), University of Padua, Padua 35131, Italy
| | - Xuchen Zhang
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mario Strazzabosco
- Liver Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
41
|
Sharma V, Letson J, Furuta S. Fibrous stroma: Driver and passenger in cancer development. Sci Signal 2022; 15:eabg3449. [PMID: 35258999 DOI: 10.1126/scisignal.abg3449] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cumulative evidence shows that fibrogenic stroma and stiff extracellular matrix (ECM) not only result from tumor growth but also play pivotal roles in cellular transformation and tumor initiation. This emerging concept may largely account for the increased cancer risk associated with environmental fibrogenic agents, such as asbestos and silica, and with chronic conditions that are fibrogenic, such as obesity and diabetes. It may also contribute to poor outcomes in patients treated with certain chemotherapeutics that can promote fibrosis, such as bleomycin and methotrexate. Although the mechanistic details of this phenomenon are still being unraveled, we provide an overview of the experimental evidence linking fibrogenic stroma and tumor initiation. In this Review, we will summarize the causes and consequences of fibrous stroma and how this stromal cue is transmitted to the nuclei of parenchymal cells through a physical continuum from the ECM to chromatin, as well as ECM-dependent biochemical signaling that contributes to cellular transformation.
Collapse
Affiliation(s)
- Vandana Sharma
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA
| | - Joshua Letson
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA
| | - Saori Furuta
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA
| |
Collapse
|
42
|
Faraldo MM, Glukhova MA, Deugnier MA. Orthotopic Transplantation of Mouse Mammary Epithelial Cells. Methods Mol Biol 2022; 2471:123-139. [PMID: 35175594 DOI: 10.1007/978-1-0716-2193-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The orthotopic transplantation assay has provided important insights into mammary development, stem cell function, and tumorigenesis. Technically, it consists in grafting mammary tissue fragments, organoids, mammospheres, or isolated cells into the fat pads of prepubertal mice from which the endogenous epithelium has been surgically removed, thereby allowing growth and differentiation of mammary epithelial cells in their physiological environment. Here, we describe how is conducted transplantation of epithelial fragments and cells isolated from mouse mammary glands, report the various approaches currently used to evaluate the regeneration and self-renewal properties of mammary stem cells, and highlight the strengths and limitations of this in vivo grafting assay.
Collapse
Affiliation(s)
- Marisa M Faraldo
- Institut Curie, PSL Research University, CNRS UMR3215/INSERM U934, Paris, France.
| | - Marina A Glukhova
- Institut Curie, PSL Research University, CNRS, UMR144, Paris, France
| | | |
Collapse
|
43
|
CaMKII Mediates TGFβ1-Induced Fibroblasts Activation and Its Cross Talk with Colon Cancer Cells. Dig Dis Sci 2022; 67:134-145. [PMID: 33528688 DOI: 10.1007/s10620-021-06847-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/12/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Cancer-associated fibroblasts (CAFs), as the activated fibroblasts in tumor stroma, are important modifiers of tumor progression. TGFβ1 has been the mostly accepted factor to fuel normal fibroblasts transformation into CAFs. Ca2+/calmodulin-dependent protein kinase II (CaMKII) is thought to play an important role in fibroblasts activation induced by TGFβ1. The aim of this study is to investigate the potential role of CaMKII in TGFβ1-induced fibroblasts activation and CAF-like differentiation. Cross talk between CaMKII-dependent fibroblasts and colon cancer in colon cancer progression also was addressed RESULTS: Immunostaining demonstrated that in colon cancer stroma, CaMKII overexpressed in stromal CAFs. In vitro, TGFβ1 increased CAF markers expression in human colon fibroblasts CCD-18Co, but not in CaMKII depletion fibroblasts. CaMKII knockdown by CaMKII shRNA significantly inhibited TGFβ1-induced fibroblasts activation and CAF-like differentiation. Smad3, AKT, and MAPK were targeted in TGFβ1-CaMKII-mediated pathway. Human colon cancer cell line HCT-116 activated fibroblasts directly, whereas CaMKII depletion dragged CCD-18Co fibroblasts undergoing CAF-associated trans-differentiation. Furthermore, increased proliferation, migration, and invasion of colon cancer cells were stimulated when co-cultured with normal fibroblasts, but not with CaMKII depletion fibroblasts. CONCLUSIONS These findings provide evidence that CaMKII is a critical mediator in TGFβ1-induced fibroblasts activation and is involved in the cross talk with colon cancer cells. CaMKII is a potentially effective target for future treatment of colon cancer.
Collapse
|
44
|
ROS Pleiotropy in Melanoma and Local Therapy with Physical Modalities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6816214. [PMID: 34777692 PMCID: PMC8580636 DOI: 10.1155/2021/6816214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/06/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Metabolic energy production naturally generates unwanted products such as reactive oxygen species (ROS), causing oxidative damage. Oxidative damage has been linked to several pathologies, including diabetes, premature aging, neurodegenerative diseases, and cancer. ROS were therefore originally anticipated as an imperative evil, a product of an imperfect system. More recently, however, the role of ROS in signaling and tumor treatment is increasingly acknowledged. This review addresses the main types, sources, and pathways of ROS in melanoma by linking their pleiotropic roles in antioxidant and oxidant regulation, hypoxia, metabolism, and cell death. In addition, the implications of ROS in various physical therapy modalities targeting melanoma, such as radiotherapy, electrochemotherapy, hyperthermia, photodynamic therapy, and medical gas plasma, are also discussed. By including ROS in the main picture of melanoma skin cancer and as an integral part of cancer therapies, a greater understanding of melanoma cell biology is presented, which ultimately may elucidate additional clues on targeting therapy resistance of this most deadly form of skin cancer.
Collapse
|
45
|
Becker L, Janssen N, Layland SL, Mürdter TE, Nies AT, Schenke-Layland K, Marzi J. Raman Imaging and Fluorescence Lifetime Imaging Microscopy for Diagnosis of Cancer State and Metabolic Monitoring. Cancers (Basel) 2021; 13:cancers13225682. [PMID: 34830837 PMCID: PMC8616063 DOI: 10.3390/cancers13225682] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Hurdles for effective tumor therapy are delayed detection and limited effectiveness of systemic drug therapies by patient-specific multidrug resistance. Non-invasive bioimaging tools such as fluorescence lifetime imaging microscopy (FLIM) and Raman-microspectroscopy have evolved over the last decade, providing the potential to be translated into clinics for early-stage disease detection, in vitro drug screening, and drug efficacy studies in personalized medicine. Accessing tissue- and cell-specific spectral signatures, Raman microspectroscopy has emerged as a diagnostic tool to identify precancerous lesions, cancer stages, or cell malignancy. In vivo Raman measurements have been enabled by recent technological advances in Raman endoscopy and signal-enhancing setups such as coherent anti-stokes Raman spectroscopy or surface-enhanced Raman spectroscopy. FLIM enables in situ investigations of metabolic processes such as glycolysis, oxidative stress, or mitochondrial activity by using the autofluorescence of co-enzymes NADH and FAD, which are associated with intrinsic proteins as a direct measure of tumor metabolism, cell death stages and drug efficacy. The combination of non-invasive and molecular-sensitive in situ techniques and advanced 3D tumor models such as patient-derived organoids or microtumors allows the recapitulation of tumor physiology and metabolism in vitro and facilitates the screening for patient-individualized drug treatment options.
Collapse
Affiliation(s)
- Lucas Becker
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
| | - Nicole Janssen
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Shannon L Layland
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
| | - Thomas E Mürdter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Anne T Nies
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tübingen, 72076 Tübingen, Germany
| | - Katja Schenke-Layland
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
- Cardiovascular Research Laboratories, Department of Medicine/Cardiology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90073, USA
| | - Julia Marzi
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| |
Collapse
|
46
|
Xu H, Pan Y. A prognostic fibroblast-related risk signature in colorectal cancer. Aging (Albany NY) 2021; 13:24251-24270. [PMID: 34735373 PMCID: PMC8610139 DOI: 10.18632/aging.203677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/26/2021] [Indexed: 11/25/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world. The accessibility of the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus data allows the prognostic evaluation of CRC. Fibroblasts play a key role in the development and progression of tumors while fibroblast-related risk signature in CRC patients has rarely been mentioned. In this study, TCGA data was classified into high-fibroblast and low-fibroblast groups according to the median of fibroblast content. Among 3845 differentially expressed genes between two groups, 14 prognostic genes commonly expressed in GSE39582 and TCGA were identified by LASSO-COX analysis. Then we established a fibroblast-related risk signature in TCGA training group and validated in the GSE39582 testing group. The risk score was significantly associated with the overall survival (OS), and the poor prognosis of patients in high-risk group might relate to the immune cell infiltration in the tumor microenvironment, epithelial-mesenchymal transition, and extracellular matrix related processes. Overall, we proved that the fibroblast-related signature could predict the prognosis of patients which might shed light on the treatment of CRC.
Collapse
Affiliation(s)
- Hao Xu
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing 100034, People's Republic of China.,Translational Cancer Research Center, Peking University First Hospital, Peking University, Beijing 100034, People's Republic of China
| | - Yisheng Pan
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing 100034, People's Republic of China
| |
Collapse
|
47
|
McGovern JA, Bock N, Shafiee A, Martine LC, Wagner F, Baldwin JG, Landgraf M, Lahr CA, Meinert C, Williams ED, Pollock PM, Denham J, Russell PJ, Risbridger GP, Clements JA, Loessner D, Holzapfel BM, Hutmacher DW. A humanized orthotopic tumor microenvironment alters the bone metastatic tropism of prostate cancer cells. Commun Biol 2021; 4:1014. [PMID: 34462519 PMCID: PMC8405640 DOI: 10.1038/s42003-021-02527-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 07/25/2021] [Indexed: 01/14/2023] Open
Abstract
Prostate cancer (PCa) is the second most commonly diagnosed cancer in men, and bone is the most frequent site of metastasis. The tumor microenvironment (TME) impacts tumor growth and metastasis, yet the role of the TME in PCa metastasis to bone is not fully understood. We used a tissue-engineered xenograft approach in NOD-scid IL2Rγnull (NSG) mice to incorporate two levels of humanization; the primary tumor and TME, and the secondary metastatic bone organ. Bioluminescent imaging, histology, and immunohistochemistry were used to study metastasis of human PC-3 and LNCaP PCa cells from the prostate to tissue-engineered bone. Here we show pre-seeding scaffolds with human osteoblasts increases the human cellular and extracellular matrix content of bone constructs, compared to unseeded scaffolds. The humanized prostate TME showed a trend to decrease metastasis of PC-3 PCa cells to the tissue-engineered bone, but did not affect the metastatic potential of PCa cells to the endogenous murine bones or organs. On the other hand, the humanized TME enhanced LNCaP tumor growth and metastasis to humanized and murine bone. Together this demonstrates the importance of the TME in PCa bone tropism, although further investigations are needed to delineate specific roles of the TME components in this context.
Collapse
Affiliation(s)
- Jacqui A McGovern
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,School of Mechanical, Medical and Process Engineering (MMPE), Centre for Biomedical Technologies, Faculty of Engineering, QUT, Brisbane, QLD, Australia.,School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Nathalie Bock
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, QUT, Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia
| | - Abbas Shafiee
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia.,Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD, Australia
| | - Laure C Martine
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Ferdinand Wagner
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Musculoskeletal University Centre Munich, Department of Orthopedics and Trauma Surgery, University Hospital Munich, Ludwig-Maximilians University, Campus Großhadern, Munich, Germany.,Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Jeremy G Baldwin
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Marietta Landgraf
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Christoph A Lahr
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,School of Mechanical, Medical and Process Engineering (MMPE), Centre for Biomedical Technologies, Faculty of Engineering, QUT, Brisbane, QLD, Australia.,Musculoskeletal University Centre Munich, Department of Orthopedics and Trauma Surgery, University Hospital Munich, Ludwig-Maximilians University, Campus Großhadern, Munich, Germany
| | - Christoph Meinert
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD, Australia
| | - Elizabeth D Williams
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, QUT, Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
| | - Pamela M Pollock
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Jim Denham
- School of Medicine and Population Health, University of Newcastle, Callaghan, NSW, Australia
| | - Pamela J Russell
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, QUT, Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Judith A Clements
- School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, QUT, Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia
| | - Daniela Loessner
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia.,Department of Anatomy and Developmental Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.,Department of Chemical Engineering and Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Melbourne, VIC, Australia
| | - Boris M Holzapfel
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia.,Musculoskeletal University Centre Munich, Department of Orthopedics and Trauma Surgery, University Hospital Munich, Ludwig-Maximilians University, Campus Großhadern, Munich, Germany
| | - Dietmar W Hutmacher
- Centre in Regenerative Medicine, Queensland University of Technology (QUT), Brisbane, QLD, Australia. .,School of Mechanical, Medical and Process Engineering (MMPE), Centre for Biomedical Technologies, Faculty of Engineering, QUT, Brisbane, QLD, Australia. .,School of Biomedical Sciences at Translational Research Institute (TRI), Faculty of Health, QUT, Brisbane, QLD, Australia. .,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia. .,ARC Industrial Transformation Training Centre in Additive Biomanufacturing, QUT, Brisbane, QLD, Australia.
| |
Collapse
|
48
|
Chuang HL, Chang YC, Huang YT, Liao JW, Kao PL, Chen YF, Lin BY, Lin YL, Chen TH, Wang YC. Establishment and Characterization of Feline Mammary Tumor Patient-Derived Xenograft Model. Animals (Basel) 2021; 11:ani11082380. [PMID: 34438836 PMCID: PMC8388766 DOI: 10.3390/ani11082380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Feline mammary tumor (FMT) is a relatively commonly diagnosed neoplasm in the cat. Development of new veterinary cancer therapies is limited by the shortage of in vivo models that reproduce tumor microenvironment and metastatic progression. In this study, FMT patient-derived xenografts (PDX) model were established and primary cell line isolated from orthotopic PDX. The tumor grafts conserve original tumor essential features, including distant metastasis. FMT-PDX represents an available resource for bridging the biology of FMT with preclinical studies of FMT in cats. Abstract Feline mammary tumor is a relatively commonly diagnosed neoplasm in the cat. Development of new veterinary cancer therapies is limited by the shortage of in vivo models that reproduce tumor microenvironment and metastatic progression. Four feline mammary tumor orthotopic patient-derived xenograft model (PDX) successfully established in NOD-SCID gamma (NSG) mice. The overall success rate of PDX establishment was 36% (4/11). Histological, immunohistochemical, and short tandem repeat analysis showed a remarkable similarity between patient’s tumor and xenograft. The tumor grafts conserve original tumor essential features, including distant metastasis. Primary FMT-1807 cell line isolated from FMT-1807PDX tumor tissue. Tumorigenicity of FMT-1807 cells expanded from PDX was assessed by orthotopic injection into NSG mice. Mice yielded tumors which preserve the lung and liver metastasis ability. This work provides a platform for FMT translational investigation.
Collapse
Affiliation(s)
- Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei 106214, Taiwan;
| | - Yi-Chih Chang
- Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan;
| | - Yi-Ting Huang
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung 402, Taiwan; (Y.-T.H.); (J.-W.L.); (P.-L.K.); (Y.-F.C.); (B.-Y.L.); (Y.-L.L.); (T.-H.C.)
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung 402, Taiwan; (Y.-T.H.); (J.-W.L.); (P.-L.K.); (Y.-F.C.); (B.-Y.L.); (Y.-L.L.); (T.-H.C.)
| | - Pei-Ling Kao
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung 402, Taiwan; (Y.-T.H.); (J.-W.L.); (P.-L.K.); (Y.-F.C.); (B.-Y.L.); (Y.-L.L.); (T.-H.C.)
| | - Yi-Fei Chen
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung 402, Taiwan; (Y.-T.H.); (J.-W.L.); (P.-L.K.); (Y.-F.C.); (B.-Y.L.); (Y.-L.L.); (T.-H.C.)
| | - Bin-Yin Lin
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung 402, Taiwan; (Y.-T.H.); (J.-W.L.); (P.-L.K.); (Y.-F.C.); (B.-Y.L.); (Y.-L.L.); (T.-H.C.)
| | - Yi-Lo Lin
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung 402, Taiwan; (Y.-T.H.); (J.-W.L.); (P.-L.K.); (Y.-F.C.); (B.-Y.L.); (Y.-L.L.); (T.-H.C.)
| | - Ter-Hsin Chen
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung 402, Taiwan; (Y.-T.H.); (J.-W.L.); (P.-L.K.); (Y.-F.C.); (B.-Y.L.); (Y.-L.L.); (T.-H.C.)
| | - Yu-Chih Wang
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung 402, Taiwan; (Y.-T.H.); (J.-W.L.); (P.-L.K.); (Y.-F.C.); (B.-Y.L.); (Y.-L.L.); (T.-H.C.)
- Correspondence: ; Tel.: +886-4-2284-0368 (ext. 92)
| |
Collapse
|
49
|
Xu Y, Liu Y, He T, Zhang Y, Wang M, Yuan H, Yang M. Biguanides decorated albumin nanoparticles loading nintedanib for synergic enhanced hepatocellular carcinoma therapy. Colloids Surf B Biointerfaces 2021; 207:112020. [PMID: 34403984 DOI: 10.1016/j.colsurfb.2021.112020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/29/2021] [Accepted: 08/01/2021] [Indexed: 12/12/2022]
Abstract
Nintedanib (ND) was known as a triple tyrosine kinase inhibitors, inhibiting angiogenesis and tissue fibrosis. Biguanide group has attracted much attention for its great penetrating ability to the lipid bilayer of cytomembrane and potential anti-cancer efficacy. In this study, a biguanide group (p-biguanidinobenzoic acid, CBH) decorated bovine serum albumin (CBH-AB) was synthesized as a novel functional biomaterial to prepare the ND loaded CBH-AB nanoparticles (ND-CBH-AB NPs). The results of physical and chemical properties showed that ND-CBH-AB NPs possessed high encapsulation efficiency and drug loading efficiency. In vitro cell study indicated that CBH modification on ND-CBH-AB NPs enhanced the cyctotoxicities to HepG2 cells. Furthermore, pharmacokinetic study showed that ND-CBH-AB NPs had good stability in circulation. Finally, pharmacodynamic studies were conducted, the results indicated that ND-CBH-AB NPs exhibited excellent anti-tumor effect and tumor microenvironment regulation effect. Thereby, this work provides a potential function albumin delivery system for hepatocellular carcinoma therapy.
Collapse
Affiliation(s)
- Ying Xu
- College of Pharmacy, Jiangsu University, Zhenjiang, 212013, China.
| | - Yulong Liu
- College of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Taofeng He
- College of Pharmacy, Jiangsu University, Zhenjiang, 212013, China; Humanwell PuraCap Pharmaceuticals (Wuhan) Co., Ltd, Wuhan, 430206, China
| | - Yaqi Zhang
- College of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Mingyun Wang
- Cancer Center of NanJing GaoChun People's Hospital, Nanjing, 211300, China
| | - Huaqin Yuan
- Cancer Center of NanJing GaoChun People's Hospital, Nanjing, 211300, China
| | - Mi Yang
- Cancer Center of NanJing GaoChun People's Hospital, Nanjing, 211300, China; The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
50
|
Rudnick JA, Monkkonen T, Mar FA, Barnes JM, Starobinets H, Goldsmith J, Roy S, Bustamante Eguiguren S, Weaver VM, Debnath J. Autophagy in stromal fibroblasts promotes tumor desmoplasia and mammary tumorigenesis. Genes Dev 2021; 35:963-975. [PMID: 34168038 PMCID: PMC8247603 DOI: 10.1101/gad.345629.120] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 05/24/2021] [Indexed: 01/11/2023]
Abstract
Autophagy inhibitors are currently being evaluated in clinical trials for the treatment of diverse cancers, largely due to their ability to impede tumor cell survival and metabolic adaptation. More recently, there is growing interest in whether and how modulating autophagy in the host stroma influences tumorigenesis. Fibroblasts play prominent roles in cancer initiation and progression, including depositing type 1 collagen and other extracellular matrix (ECM) components, thereby stiffening the surrounding tissue to enhance tumor cell proliferation and survival, as well as secreting cytokines that modulate angiogenesis and the immune microenvironment. This constellation of phenotypes, pathologically termed desmoplasia, heralds poor prognosis and reduces patient survival. Using mouse mammary cancer models and syngeneic transplantation assays, we demonstrate that genetic ablation of stromal fibroblast autophagy significantly impedes fundamental elements of the stromal desmoplastic response, including collagen and proinflammatory cytokine secretion, extracellular matrix stiffening, and neoangiogenesis. As a result, autophagy in stromal fibroblasts is required for mammary tumor growth in vivo, even when the cancer cells themselves remain autophagy-competent . We propose the efficacy of autophagy inhibition is shaped by this ability of host stromal fibroblast autophagy to support tumor desmoplasia.
Collapse
Affiliation(s)
- Jenny A Rudnick
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA
| | - Teresa Monkkonen
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA
| | - Florie A Mar
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA
- Biomedical Sciences Graduate Program, University of California at San Francisco, San Francisco, California 94143, USA
| | - James M Barnes
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California at San Francisco, San Francisco, California 94143, USA
| | - Hanna Starobinets
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA
- Biomedical Sciences Graduate Program, University of California at San Francisco, San Francisco, California 94143, USA
| | - Juliet Goldsmith
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA
- Biomedical Sciences Graduate Program, University of California at San Francisco, San Francisco, California 94143, USA
| | - Srirupa Roy
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA
| | - Sofía Bustamante Eguiguren
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California at San Francisco, San Francisco, California 94143, USA
- Department of Anatomy, University of California at San Francisco, San Francisco, California 94143, USA
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, California 94143, USA
| | - Jayanta Debnath
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA
| |
Collapse
|