1
|
DU N, Wan H, Guo H, Zhang X, Wu X. [Myeloid-derived suppressor cells as important factors and potential targets for breast cancer progression]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:785-795. [PMID: 39686697 DOI: 10.3724/zdxbyxb-2024-0353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Recurrence and metastasis remain the leading cause of death in breast cancer patients due to the lack of effective treatment. A microenvironment suitable for cancer cell growth, referred to as pre-metastatic niche (PMN), is formed in distant organs before metastasis occurs. Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of immature myeloid cells with immunosuppressive effects. They can expand in large numbers in breast cancer patients and participate in the formation of PMN. MDSCs can remodel the extracellular matrix of pulmonary vascular endothelial cells and recruit cancer stem cells to promote the lung metastasis of breast cancer. Furthermore, MDSCs facilitate immune evasion of breast cancer cells to impact the efficacy of immunotherapy. It is proposed that MDSCs represent a potential therapeutic target for the inhibition of recurrence and metastasis in breast cancer. Therapeutic strategies targeting MDSCs have shown promising efficacy in preclinical studies and clinical trials. This review presents a summary of the principal factors involved in the recruitment and activation of MDSCs during the formation of PMN, and outlines MDSCs functions such as immunosuppression and the current targeted therapies against MDSCs, aiming to provide new ideas for the treatment of distant metastases in breast cancer.
Collapse
Affiliation(s)
- Nannan DU
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China.
| | - Hua Wan
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Hailing Guo
- Department of Orthopaedics and Traumatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Xukuan Zhang
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Xueqing Wu
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China.
| |
Collapse
|
2
|
Serrano García L, Jávega B, Llombart Cussac A, Gión M, Pérez-García JM, Cortés J, Fernández-Murga ML. Patterns of immune evasion in triple-negative breast cancer and new potential therapeutic targets: a review. Front Immunol 2024; 15:1513421. [PMID: 39735530 PMCID: PMC11671371 DOI: 10.3389/fimmu.2024.1513421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by the absence of progesterone and estrogen receptors and low (or absent) HER2 expression. TNBC accounts for 15-20% of all breast cancers. It is associated with younger age, a higher mutational burden, and an increased risk of recurrence and mortality. Standard treatment for TNBC primarily relies on cytotoxic agents, such as taxanes, anthracyclines, and platinum compounds for both early and advanced stages of the disease. Several targeted therapies, including bevacizumab and sunitinib, have failed to demonstrate significant clinical benefit in TNBC. The emergence of immune checkpoint inhibitors (ICI) has revolutionized cancer treatment. By stimulating the immune system, ICIs induce a durable anti-tumor response across various solid tumors. TNBC is a particularly promising target for treatment with ICIs due to the higher levels of tumor-infiltrating lymphocytes (TIL), increased PD-L1 expression, and higher mutational burden, which generates tumor-specific neoantigens that activate immune cells. ICIs administered as monotherapy in advanced TNBC yields only a modest response; however, response rates significantly improve when ICIs are combined with cytotoxic agents, particularly in tumors expressing PD-L1. Pembrolizumab is approved for use in both early and advanced TNBC in combination with standard chemotherapy. However, more research is needed to identify more potent biomarkers, and to better elucidate the synergism of ICIs with other targeted agents. In this review, we explore the challenges of immunotherapy in TNBC, examining the mechanisms of tumor progression mediated by immune cells within the tumor microenvironment, and the signaling pathways involved in both primary and acquired resistance. Finally, we provide a comprehensive overview of ongoing clinical trials underway to investigate novel immune-targeted therapies for TNBC.
Collapse
Affiliation(s)
- Lucía Serrano García
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
| | - Beatriz Jávega
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
| | - Antonio Llombart Cussac
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
- Grupo Oncología Traslacional, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-Centro de Estudios Universitarios (CEU), Alfara del Patriarca, Spain
- Medica Scientia Innovation Research (MEDSIR), Oncoclínicas & Co., Jersey City, NJ, United States
| | - María Gión
- Medical Oncology Department, Hospital Ramon y Cajal, Madrid, Spain
| | - José Manuel Pérez-García
- Medica Scientia Innovation Research (MEDSIR), Oncoclínicas & Co., Jersey City, NJ, United States
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
| | - Javier Cortés
- Medica Scientia Innovation Research (MEDSIR), Oncoclínicas & Co., Jersey City, NJ, United States
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
- Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
| | - María Leonor Fernández-Murga
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
| |
Collapse
|
3
|
Mao C, Chen Y, Xing D, Zhang T, Lin Y, Long C, Yu J, Luo Y, Ming T, Xie W, Han Z, Mei D, Xiang D, Lu M, Shen X, Xue X. Resting natural killer cells promote the progress of colon cancer liver metastasis by elevating tumor-derived stem cell factor. eLife 2024; 13:RP97201. [PMID: 39387546 PMCID: PMC11466454 DOI: 10.7554/elife.97201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2024] Open
Abstract
The abundance and biological contribution of natural killer (NK) cells in cancer are controversial. Here, we aim to uncover clinical relevance and cellular roles of NK cells in colon cancer liver metastasis (CCLM). Here, we integrated single-cell RNA-sequencing, spatial transcriptomics (ST), and bulk RNA-sequencing datasets to investigate NK cells' biological properties and functions in the microenvironment of primary and liver metastatic tumors. Results were validated through an in vitro co-culture experiment based on bioinformatics analysis. Useing single-cell RNA-sequencing and ST, we mapped the immune cellular landscape of colon cancer and well-matched liver metastatic cancer. We discovered that GZMK+ resting NK cells increased significantly in tumor tissues and were enriched in the tumor regions of both diseases. After combining bulk RNA and clinical data, we observed that these NK cell subsets contributed to a worse prognosis. Meanwhile, KIR2DL4+ activated NK cells exhibited the opposite position and relevance. Pseudotime cell trajectory analysis revealed the evolution of activated to resting NK cells. In vitro experiments further confirmed that tumor-cell-co-cultured NK cells exhibited a decidual-like status, as evidenced by remarkable increasing CD9 expression. Functional experiments finally revealed that NK cells exhibited tumor-activating characteristics by promoting the dissociation of SCF (stem cell factor) on the tumor cells membrane depending on cell-to-cell interaction, as the supernatant of the co-culture system enhanced tumor progression. In summary, our findings revealed resting NK cells exhibited a clinical relevance with CCLM, which may be exploited for novel strategies to improve therapeutic outcomes for patients with CCLM.
Collapse
Affiliation(s)
- Chenchen Mao
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Yanyu Chen
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
- Department of Pediatric Thoracic Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Dong Xing
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Teming Zhang
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Yangxuan Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Cong Long
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Jiaye Yu
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Yunhui Luo
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Tao Ming
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Wangkai Xie
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Zheng Han
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Dianfeng Mei
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Dan Xiang
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| | - Mingdong Lu
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xian Shen
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiangyang Xue
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
4
|
Sarangi P. Role of indoleamine 2, 3-dioxygenase 1 in immunosuppression of breast cancer. CANCER PATHOGENESIS AND THERAPY 2024; 2:246-255. [PMID: 39371092 PMCID: PMC11447360 DOI: 10.1016/j.cpt.2023.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/27/2023] [Accepted: 11/03/2023] [Indexed: 10/08/2024]
Abstract
Breast cancer (BC) contributes greatly to global cancer incidence and is the main cause of cancer-related deaths among women globally. It is a complex disease characterized by numerous subtypes with distinct clinical manifestations. Immune checkpoint inhibitors (ICIs) are not effective in all patients and have been associated with tumor resistance and immunosuppression. Because amino acid (AA)-catabolizing enzymes have been shown to regulate immunosuppressive effects, this review investigated the immunosuppressive roles of indoleamine 2,3-dioxygenase 1 (IDO1), a tryptophan (Trp)-catabolizing enzyme, which is overexpressed in various metastatic tumors. It promotes immunomodulatory effects by depleting Trp in the regional microenvironment. This leads to a reduction in the number of immunogenic immune cells, such as effector T and natural killer (NK) cells, and an increase in tolerogenic immune cells, such as regulatory T (Treg) cells. The BC tumor microenvironment (TME) establishes a supportive niche where cancer cells can interact with immune cells and neighboring endothelial cells and is thus a feasible target for cancer therapy. In many immunological contexts, IDO1 regulates immune control by causing regional metabolic changes in the TME and tissue environment, which may further affect the maturation of systemic immunological tolerance. In the development of effective treatment targets and approaches, it is essential to understand the immunomodulatory effects exerted by AA-catabolizing enzymes, such as IDO1, on the components of the TME.
Collapse
Affiliation(s)
- Pratyasha Sarangi
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha 751024, India
| |
Collapse
|
5
|
Doran BR, Moffitt LR, Wilson AL, Stephens AN, Bilandzic M. Leader Cells: Invade and Evade-The Frontline of Cancer Progression. Int J Mol Sci 2024; 25:10554. [PMID: 39408880 PMCID: PMC11476628 DOI: 10.3390/ijms251910554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Metastasis is the leading cause of cancer-related mortality; however, a complete understanding of the molecular programs driving the metastatic cascade is lacking. Metastasis is dependent on collective invasion-a developmental process exploited by many epithelial cancers to establish secondary tumours and promote widespread disease. The key drivers of collective invasion are "Leader Cells", a functionally distinct subpopulation of cells that direct migration, cellular contractility, and lead trailing or follower cells. While a significant body of research has focused on leader cell biology in the traditional context of collective invasion, the influence of metastasis-promoting leader cells is an emerging area of study. This review provides insights into the expanded role of leader cells, detailing emerging evidence on the hybrid epithelial-mesenchymal transition (EMT) state and the phenotypical plasticity exhibited by leader cells. Additionally, we explore the role of leader cells in chemotherapeutic resistance and immune evasion, highlighting their potential as effective and diverse targets for novel cancer therapies.
Collapse
Affiliation(s)
- Brittany R. Doran
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Laura R. Moffitt
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Amy L. Wilson
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Andrew N. Stephens
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research, Clayton 3168, Australia; (B.R.D.); (L.R.M.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| |
Collapse
|
6
|
Wang Z, Sha T, Li J, Luo H, Liu A, Liang H, Qiang J, Li L, Whittaker AK, Yang B, Sun H, Shi C, Lin Q. Turning foes to friends: Advanced " in situ nanovaccine" with dual immunoregulation for enhanced immunotherapy of metastatic triple-negative breast cancer. Bioact Mater 2024; 39:612-629. [PMID: 38883315 PMCID: PMC11179173 DOI: 10.1016/j.bioactmat.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/21/2024] [Accepted: 04/21/2024] [Indexed: 06/18/2024] Open
Abstract
As a "cold tumor", triple-negative breast cancer (TNBC) exhibits limited responsiveness to current immunotherapy. How to enhance the immunogenicity and reverse the immunosuppressive microenvironment of TNBC remain a formidable challenge. Herein, an "in situ nanovaccine" Au/CuNDs-R848 was designed for imaging-guided photothermal therapy (PTT)/chemodynamic therapy (CDT) synergistic therapy to trigger dual immunoregulatory effects on TNBC. On the one hand, Au/CuNDs-R848 served as a promising photothermal agent and nanozyme, achieving PTT and photothermal-enhanced CDT against the primary tumor of TNBC. Meanwhile, the released antigens and damage-associated molecular patterns (DAMPs) promoted the maturation of dendritic cells (DCs) and facilitated the infiltration of T lymphocytes. Thus, Au/CuNDs-R848 played a role as an "in situ nanovaccine" to enhance the immunogenicity of TNBC by inducing immunogenic cell death (ICD). On the other hand, the nanovaccine suppressed the myeloid-derived suppressor cells (MDSCs), thereby reversing the immunosuppressive microenvironment. Through the dual immunoregulation, "cold tumor" was transformed into a "hot tumor", not only implementing a "turning foes to friends" therapeutic strategy but also enhancing immunotherapy against metastatic TNBC. Furthermore, Au/CuNDs-R848 acted as an excellent nanoprobe, enabling high-resolution near-infrared fluorescence and computed tomography imaging for precise visualization of TNBC. This feature offers potential applications in clinical cancer detection and surgical guidance. Collectively, this work provides an effective strategy for enhancing immune response and offers novel insights into the potential clinical applications for tumor immunotherapy.
Collapse
Affiliation(s)
- Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Tong Sha
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Jinwei Li
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Huanyu Luo
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Annan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Hao Liang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Jinbiao Qiang
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Lei Li
- Department of Endocrinology, Lequn Branch, The First Hospital of Jilin University, 130021, PR China
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Hongchen Sun
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Ce Shi
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| |
Collapse
|
7
|
Hao X, Jiang B, Wu J, Xiang D, Xiong Z, Li C, Li Z, He S, Tu C, Li Z. Nanomaterials for bone metastasis. J Control Release 2024; 373:640-651. [PMID: 39084467 DOI: 10.1016/j.jconrel.2024.07.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
Bone metastasis, a prevalent occurrence in primary malignant tumors, is often associated with a grim prognosis. The bone microenvironment comprises various coexisting cell types, working together in a coordinated manner. This dynamic microenvironment plays a pivotal role in the initiation and progression of bone metastases. While cancer therapies have made advancements, the available options for addressing bone metastases remain insufficient. The advent of nanotechnology has ushered in a new era for managing and preventing bone metastases because of the physicochemical and adaptable advantages of nanoplatforms. In this review, we make an introduction of the underlying mechanisms and the current clinical therapies of bone metastases, highlighting the advances of intelligent nanosystems that can stimulate vascular regeneration, promote bone regeneration, eliminate tumor cells, minimize bone damage, and expedite bone healing. The innovation surrounding bone-targeting nanoplatforms presents a fresh approach to the theranostics of bone metastases.
Collapse
Affiliation(s)
- Xinyan Hao
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410011, China; Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Buchan Jiang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zijian Xiong
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Chenbei Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhaoqi Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Shasha He
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Changsha Medical University, Changsha 410219, China.
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Shenzhen Research Institute of Central South University, Guangdong 518063, China; FuRong Laboratory, Changsha 410078, Hunan, China.
| |
Collapse
|
8
|
Kitelinger LE, Thim EA, Zipkowitz SY, Price RJ, Bullock TNJ. Tissue- and Temporal-Dependent Dynamics of Myeloablation in Response to Gemcitabine Chemotherapy. Cells 2024; 13:1317. [PMID: 39195207 DOI: 10.3390/cells13161317] [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: 06/15/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/29/2024] Open
Abstract
For triple-negative breast cancer (TNBC), the most aggressive subset of breast cancer, immune cell infiltrates have prognostic implications. The presence of myeloid-derived suppressor cells supports tumor progression, while tumor-infiltrating lymphocytes (TILs) correlate with improved survival and responsiveness to immunotherapy. Manipulating the abundance of these populations may enhance tumor immunity. Gemcitabine (GEM), a clinically employed chemotherapeutic, is reported to be systemically myeloablative, and thus it is a potentially useful adjunct therapy for promoting anti-tumor immunity. However, knowledge about the immunological effects of GEM intratumorally is limited. Thus, we directly compared the impact of systemic GEM on immune cell presence and functionality in the tumor microenvironment (TME) to its effects in the periphery. We found that GEM is not myeloablative in the TME; rather, we observed sustained, significant reductions in TILs and dendritic cells-crucial components in initiating an adaptive immune response. We also performed bulk-RNA sequencing to identify immunological alterations transcriptionally induced by GEM. While we found evidence of upregulation in the interferon-gamma (IFN-γ) response pathway, we determined that GEM-mediated growth control is not dependent on IFN-γ. Overall, our findings yield new insights into the tissue- and temporal-dependent immune ablative effects of GEM, contrasting the paradigm that this therapy is specifically myeloablative.
Collapse
Affiliation(s)
- Lydia E Kitelinger
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Eric A Thim
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Sarah Y Zipkowitz
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Timothy N J Bullock
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
9
|
Zou Z, Luo T, Wang X, Wang B, Li Q. Exploring the interplay between triple-negative breast cancer stem cells and tumor microenvironment for effective therapeutic strategies. J Cell Physiol 2024; 239:e31278. [PMID: 38807378 DOI: 10.1002/jcp.31278] [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: 02/26/2024] [Revised: 03/28/2024] [Accepted: 04/05/2024] [Indexed: 05/30/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive and metastatic malignancy with poor treatment outcomes. The interaction between the tumor microenvironment (TME) and breast cancer stem cells (BCSCs) plays an important role in the development of TNBC. Owing to their ability of self-renewal and multidirectional differentiation, BCSCs maintain tumor growth, drive metastatic colonization, and facilitate the development of drug resistance. TME is the main factor regulating the phenotype and metastasis of BCSCs. Immune cells, cancer-related fibroblasts (CAFs), cytokines, mesenchymal cells, endothelial cells, and extracellular matrix within the TME form a complex communication network, exert highly selective pressure on the tumor, and provide a conducive environment for the formation of BCSC niches. Tumor growth and metastasis can be controlled by targeting the TME to eliminate BCSC niches or targeting BCSCs to modify the TME. These approaches may improve the treatment outcomes and possess great application potential in clinical settings. In this review, we summarized the relationship between BCSCs and the progression and drug resistance of TNBC, especially focusing on the interaction between BCSCs and TME. In addition, we discussed therapeutic strategies that target the TME to inhibit or eliminate BCSCs, providing valuable insights into the clinical treatment of TNBC.
Collapse
Affiliation(s)
- Zhuoling Zou
- Queen Mary College, Nanchang University, Nanchang, Jiangxi, China
| | - Tinglan Luo
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing, China
| | - Xinyuan Wang
- Department of Clinical Medicine, The Second Clinical College of Chongqing Medicine University, Chongqing, China
| | - Bin Wang
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing, China
| | - Qing Li
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
10
|
Otterlei Fjørtoft M, Huse K, Rye IH. The Tumor Immune Microenvironment in Breast Cancer Progression. Acta Oncol 2024; 63:359-367. [PMID: 38779867 PMCID: PMC11332517 DOI: 10.2340/1651-226x.2024.33008] [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: 12/01/2023] [Accepted: 02/12/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND The tumor microenvironment significantly influences breast cancer development, progression, and metastasis. Various immune cell populations, including T cells, B cells, NK cells, and myeloid cells exhibit diverse functions in different breast cancer subtypes, contributing to both anti-tumor and pro-tumor activities. PURPOSE This review provides an overview of the predominant immune cell populations in breast cancer subtypes, elucidating their suppressive and prognostic effects. We aim to outline the role of the immune microenvironment from normal breast tissue to invasive cancer and distant metastasis. METHODS A comprehensive literature review was conducted to analyze the involvement of immune cells throughout breast cancer progression. RESULTS In breast cancer, tumors exhibit increased immune cell infiltration compared to normal tissue. Variations exist across subtypes, with higher levels observed in triple-negative and HER2+ tumors are linked to better survival. In contrast, ER+ tumors display lower immune infiltration, associated with poorer outcomes. Furthermore, metastatic sites commonly exhibit a more immunosuppressive microenvironment. CONCLUSION Understanding the complex interaction between tumor and immune cells during breast cancer progression is essential for future research and the development of immune-based strategies. This comprehensive understanding may pave the way for more effective treatment approaches and improved patients outcomes.
Collapse
Affiliation(s)
- Marit Otterlei Fjørtoft
- Department of Cancer Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Radium Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Radium Hospital, Oslo, Norway
| | - Inga Hansine Rye
- Department of Cancer Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, Radium Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
11
|
Peng X, Dong H, Zhang L, Liu S. Role of cancer stem cell ecosystem on breast cancer metastasis and related mouse models. Zool Res 2024; 45:506-517. [PMID: 38682432 PMCID: PMC11188611 DOI: 10.24272/j.issn.2095-8137.2023.411] [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: 02/20/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024] Open
Abstract
Breast cancer metastasis is responsible for most breast cancer-related deaths and is influenced by many factors within the tumor ecosystem, including tumor cells and microenvironment. Breast cancer stem cells (BCSCs) constitute a small population of cancer cells with unique characteristics, including their capacity for self-renewal and differentiation. Studies have shown that BCSCs not only drive tumorigenesis but also play a crucial role in promoting metastasis in breast cancer. The tumor microenvironment (TME), composed of stromal cells, immune cells, blood vessel cells, fibroblasts, and microbes in proximity to cancer cells, is increasingly recognized for its crosstalk with BCSCs and role in BCSC survival, growth, and dissemination, thereby influencing metastatic ability. Hence, a thorough understanding of BCSCs and the TME is critical for unraveling the mechanisms underlying breast cancer metastasis. In this review, we summarize current knowledge on the roles of BCSCs and the TME in breast cancer metastasis, as well as the underlying regulatory mechanisms. Furthermore, we provide an overview of relevant mouse models used to study breast cancer metastasis, as well as treatment strategies and clinical trials addressing BCSC-TME interactions during metastasis. Overall, this study provides valuable insights for the development of effective therapeutic strategies to reduce breast cancer metastasis.
Collapse
Affiliation(s)
- Xilei Peng
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences
- State Key Laboratory of Genetic Engineering
- Cancer Institutes
- Department of Oncology
- Key Laboratory of Breast Cancer in Shanghai
- Shanghai Key Laboratory of Medical Epigenetics
- Shanghai Key Laboratory of Radiation Oncology
- International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology
- Shanghai Medical College
- Fudan University, Shanghai 200032, China
| | - Haonan Dong
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences
- State Key Laboratory of Genetic Engineering
- Cancer Institutes
- Department of Oncology
- Key Laboratory of Breast Cancer in Shanghai
- Shanghai Key Laboratory of Medical Epigenetics
- Shanghai Key Laboratory of Radiation Oncology
- International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology
- Shanghai Medical College
- Fudan University, Shanghai 200032, China
| | - Lixing Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences
- State Key Laboratory of Genetic Engineering
- Cancer Institutes
- Department of Oncology
- Key Laboratory of Breast Cancer in Shanghai
- Shanghai Key Laboratory of Medical Epigenetics
- Shanghai Key Laboratory of Radiation Oncology
- International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology
- Shanghai Medical College
- Fudan University, Shanghai 200032, China. E-mail:
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences
- State Key Laboratory of Genetic Engineering
- Cancer Institutes
- Department of Oncology
- Key Laboratory of Breast Cancer in Shanghai
- Shanghai Key Laboratory of Medical Epigenetics
- Shanghai Key Laboratory of Radiation Oncology
- International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology
- Shanghai Medical College
- Fudan University, Shanghai 200032, China. E-mail:
| |
Collapse
|
12
|
Kim U, Debnath R, Maiz JE, Rico J, Sinha S, Blanco MA, Chakrabarti R. ΔNp63 regulates MDSC survival and metabolism in triple-negative breast cancer. iScience 2024; 27:109366. [PMID: 38510127 PMCID: PMC10951988 DOI: 10.1016/j.isci.2024.109366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/20/2023] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Triple-negative breast cancer (TNBC) contributes greatly to mortality of breast cancer, demanding new targetable options. We have shown that TNBC patients have high ΔNp63 expression in tumors. However, the function of ΔNp63 in established TNBC is yet to be explored. In current studies, targeting ΔNp63 with inducible CRISPR knockout and Histone deacetylase inhibitor Quisinostat showed that ΔNp63 is important for tumor progression and metastasis in established tumors by promoting myeloid-derived suppressor cell (MDSC) survival through tumor necrosis factor alpha. Decreasing ΔNp63 levels are associated with decreased CD4+ and FOXP3+ T-cells but increased CD8+ T-cells. RNA sequencing analysis indicates that loss of ΔNp63 alters multiple MDSC properties such as lipid metabolism, chemotaxis, migration, and neutrophil degranulation besides survival. We further demonstrated that targeting ΔNp63 sensitizes chemotherapy. Overall, we showed that ΔNp63 reprograms the MDSC-mediated immunosuppressive functions in TNBC, highlighting the benefit of targeting ΔNp63 in chemotherapy-resistant TNBC.
Collapse
Affiliation(s)
- Ukjin Kim
- Department of Surgery, Sylvester Comprehensive Cancer, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rahul Debnath
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Javier E. Maiz
- Department of Surgery, Sylvester Comprehensive Cancer, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua Rico
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Mario Andrés Blanco
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rumela Chakrabarti
- Department of Surgery, Sylvester Comprehensive Cancer, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Salembier R, De Haes C, Bellemans J, Demeyere K, Van Den Broeck W, Sanders NN, Van Laere S, Lyons TR, Meyer E, Steenbrugge J. Chitin-mediated blockade of chitinase-like proteins reduces tumor immunosuppression, inhibits lymphatic metastasis and enhances anti-PD-1 efficacy in complementary TNBC models. Breast Cancer Res 2024; 26:63. [PMID: 38605414 PMCID: PMC11007917 DOI: 10.1186/s13058-024-01815-8] [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: 08/21/2023] [Accepted: 03/23/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Chitinase-like proteins (CLPs) play a key role in immunosuppression under inflammatory conditions such as cancer. CLPs are enzymatically inactive and become neutralized upon binding of their natural ligand chitin, potentially reducing CLP-driven immunosuppression. We investigated the efficacy of chitin treatment in the context of triple-negative breast cancer (TNBC) using complementary mouse models. We also evaluated the immunomodulatory influence of chitin on immune checkpoint blockade (ICB) and compared its efficacy as general CLP blocker with blockade of a single CLP, i.e. chitinase 3-like 1 (CHI3L1). METHODS Female BALB/c mice were intraductally injected with luciferase-expressing 4T1 or 66cl4 cells and systemically treated with chitin in combination with or without anti-programmed death (PD)-1 ICB. For single CLP blockade, tumor-bearing mice were treated with anti-CHI3L1 antibodies. Metastatic progression was monitored through bioluminescence imaging. Immune cell changes in primary tumors and lymphoid organs (i.e. axillary lymph nodes and spleen) were investigated through flow cytometry, immunohistochemistry, cytokine profiling and RNA-sequencing. CHI3L1-stimulated RAW264.7 macrophages were subjected to 2D lymphatic endothelial cell adhesion and 3D lymphatic integration in vitro assays for studying macrophage-mediated lymphatic remodeling. RESULTS Chitin significantly reduced primary tumor progression in the 4T1-based model by decreasing the high production of CLPs that originate from tumor-associated neutrophils (TANs) and Stat3 signaling, prominently affecting the CHI3L1 and CHI3L3 primary tumor levels. It reduced immunosuppressive cell types and increased anti-tumorigenic T-cells in primary tumors as well as axillary lymph nodes. Chitin also significantly reduced CHI3L3 primary tumor levels and immunosuppression in the 66cl4-based model. Compared to anti-CHI3L1, chitin enhanced primary tumor growth reduction and anti-tumorigenicity. Both treatments equally inhibited lymphatic adhesion and integration of macrophages, thereby hampering lymphatic tumor cell spreading. Upon ICB combination therapy, chitin alleviated anti-PD-1 resistance in both TNBC models, providing a significant add-on reduction in primary tumor and lung metastatic growth compared to chitin monotherapy. These add-on effects occurred through additional increase in CD8α+ T-cell infiltration and activation in primary tumor and lymphoid organs. CONCLUSIONS Chitin, as a general CLP blocker, reduces CLP production, enhances anti-tumor immunity as well as ICB responses, supporting its potential clinical relevance in immunosuppressed TNBC patients.
Collapse
Affiliation(s)
- Robbe Salembier
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Caro De Haes
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Julie Bellemans
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Wim Van Den Broeck
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Niek N Sanders
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Steven Van Laere
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
14
|
Akinsipe T, Mohamedelhassan R, Akinpelu A, Pondugula SR, Mistriotis P, Avila LA, Suryawanshi A. Cellular interactions in tumor microenvironment during breast cancer progression: new frontiers and implications for novel therapeutics. Front Immunol 2024; 15:1302587. [PMID: 38533507 PMCID: PMC10963559 DOI: 10.3389/fimmu.2024.1302587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 02/16/2024] [Indexed: 03/28/2024] Open
Abstract
The breast cancer tumor microenvironment (TME) is dynamic, with various immune and non-immune cells interacting to regulate tumor progression and anti-tumor immunity. It is now evident that the cells within the TME significantly contribute to breast cancer progression and resistance to various conventional and newly developed anti-tumor therapies. Both immune and non-immune cells in the TME play critical roles in tumor onset, uncontrolled proliferation, metastasis, immune evasion, and resistance to anti-tumor therapies. Consequently, molecular and cellular components of breast TME have emerged as promising therapeutic targets for developing novel treatments. The breast TME primarily comprises cancer cells, stromal cells, vasculature, and infiltrating immune cells. Currently, numerous clinical trials targeting specific TME components of breast cancer are underway. However, the complexity of the TME and its impact on the evasion of anti-tumor immunity necessitate further research to develop novel and improved breast cancer therapies. The multifaceted nature of breast TME cells arises from their phenotypic and functional plasticity, which endows them with both pro and anti-tumor roles during tumor progression. In this review, we discuss current understanding and recent advances in the pro and anti-tumoral functions of TME cells and their implications for developing safe and effective therapies to control breast cancer progress.
Collapse
Affiliation(s)
- Tosin Akinsipe
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Rania Mohamedelhassan
- Department of Chemical Engineering, College of Engineering, Auburn University, Auburn, AL, United States
| | - Ayuba Akinpelu
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Satyanarayana R. Pondugula
- Department of Chemical Engineering, College of Engineering, Auburn University, Auburn, AL, United States
| | - Panagiotis Mistriotis
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - L. Adriana Avila
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
15
|
Said SS, Ibrahim WN. Breaking Barriers: The Promise and Challenges of Immune Checkpoint Inhibitors in Triple-Negative Breast Cancer. Biomedicines 2024; 12:369. [PMID: 38397971 PMCID: PMC10886684 DOI: 10.3390/biomedicines12020369] [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: 12/04/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 02/25/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive malignancy with pronounced immunogenicity, exhibiting rapid proliferation and immune cell infiltration into the tumor microenvironment. TNBC's heterogeneity poses challenges to immunological treatments, inducing resistance mechanisms in the tumor microenvironment. Therapeutic modalities, including immune checkpoint inhibitors (ICIs) targeting PD-1, PD-L1, and CTLA-4, are explored in preclinical and clinical trials. Promising results emerge from combining ICIs with anti-TGF-β and VISTA, hindering TNBC tumor growth. TNBC cells employ complex evasion strategies involving interactions with stromal and immune cells, suppressing immune recognition through various cytokines, chemokines, and metabolites. The recent focus on unraveling humoral and cellular components aims to disrupt cancer crosstalk within the tumor microenvironment. This review identifies TNBC's latest resistance mechanisms, exploring potential targets for clinical trials to overcome immune checkpoint resistance and enhance patient survival rates.
Collapse
Affiliation(s)
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| |
Collapse
|
16
|
Kim J, Pena JV, McQueen HP, Kong L, Michael D, Lomashvili EM, Cook PR. Downstream STING pathways IRF3 and NF-κB differentially regulate CCL22 in response to cytosolic dsDNA. Cancer Gene Ther 2024; 31:28-42. [PMID: 37990062 DOI: 10.1038/s41417-023-00678-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 08/22/2023] [Accepted: 10/11/2023] [Indexed: 11/23/2023]
Abstract
Double-stranded DNA (dsDNA) in the cytoplasm of eukaryotic cells is abnormal and typically indicates the presence of pathogens or mislocalized self-DNA. Multiple sensors detect cytosolic dsDNA and trigger robust immune responses via activation of type I interferons. Several cancer immunotherapy treatments also activate cytosolic nucleic acid sensing pathways, including oncolytic viruses, nucleic acid-based cancer vaccines, and pharmacological agonists. We report here that cytosolic dsDNA introduced into malignant cells can robustly upregulate expression of CCL22, a chemokine responsible for the recruitment of regulatory T cells (Tregs). Tregs in the tumor microenvironment are thought to repress anti-tumor immune responses and contribute to tumor immune evasion. Surprisingly, we found that CCL22 upregulation by dsDNA was mediated primarily by interferon regulatory factor 3 (IRF3), a key transcription factor that activates type I interferons. This finding was unexpected given previous reports that type I interferon alpha (IFN-α) inhibits CCL22 and that IRF3 is associated with strong anti-tumor immune responses, not Treg recruitment. We also found that CCL22 upregulation by dsDNA occurred concurrently with type I interferon beta (IFN-β) upregulation. IRF3 is one of two transcription factors downstream of the STimulator of INterferon Genes (STING), a hub adaptor protein through which multiple dsDNA sensors transmit their signals. The other transcription factor downstream of STING, NF-κB, has been reported to regulate CCL22 expression in other contexts, and NF-κB has also been associated with multiple pro-tumor functions, including Treg recruitment. However, we found that NF-κB in the context of activation by cytosolic dsDNA contributed minimally to CCL22 upregulation compared with IRF3. Lastly, we observed that two strains of the same cell line differed profoundly in their capacity to upregulate CCL22 and IFN-β in response to dsDNA, despite apparent STING activation in both cell lines. This finding suggests that during tumor evolution, cells can acquire, or lose, the ability to upregulate CCL22. This study adds to our understanding of factors that may modulate immune activation in response to cytosolic DNA and has implications for immunotherapy strategies that activate DNA sensing pathways in cancer cells.
Collapse
Affiliation(s)
- Jihyun Kim
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Jocelyn V Pena
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Hannah P McQueen
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Lingwei Kong
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Dina Michael
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Elmira M Lomashvili
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Pamela R Cook
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA, USA.
| |
Collapse
|
17
|
Samanta A, Saha P, Johnson O, Bishayee A, Sinha D. Dysregulation of delta Np63 alpha in squamous cell carcinoma and its therapeutic targeting. Biochim Biophys Acta Rev Cancer 2024; 1879:189034. [PMID: 38040268 DOI: 10.1016/j.bbcan.2023.189034] [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: 08/08/2023] [Revised: 11/05/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
The gene p63 has two isoforms -a full length transactivated isoform (TA) p63 and an amino-terminally truncated isoform, ∆Np63. DeltaNp63 alpha (∆Np63α) is the predominant splice variant of the isoform, ∆Np63 and is expressed in the basal layer of stratified epithelia. ∆Np63α that is normally essential for the epithelial lineage maintenance may be dysregulated in squamous cell carcinomas (SCCs). The pro-tumorigenic or antitumorigenic role of ∆Np63 is a highly contentious arena. ∆Np63α may act as a double-edged sword. It may either promote tumor progression, epithelial-mesenchymal transition, migration, chemoresistance, and immune-inflammatory responses, or inhibit the aforementioned phenomena depending upon cell type and tumor microenvironment. Several signaling pathways, transforming growth factor-β, Wnt and Notch, as well as epigenetic alterations involving microRNAs, and long noncoding RNAs are regulated by ∆Np63α. This review has attempted to provide an in-depth insight into the role of ∆Np63α in the development of SCCs during different stages of tumor formation and how it may be targeted for therapeutic implications.
Collapse
Affiliation(s)
- Anurima Samanta
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, West Bengal, India
| | - Priyanka Saha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, West Bengal, India
| | - Olivia Johnson
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| | - Dona Sinha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, West Bengal, India.
| |
Collapse
|
18
|
Eyermann CE, Chen X, Somuncu OS, Li J, Joukov AN, Chen J, Alexandrova EM. ΔNp63 Regulates Homeostasis, Stemness, and Suppression of Inflammation in the Adult Epidermis. J Invest Dermatol 2024; 144:73-83.e10. [PMID: 37543242 DOI: 10.1016/j.jid.2023.07.005] [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: 12/16/2022] [Revised: 06/22/2023] [Accepted: 07/10/2023] [Indexed: 08/07/2023]
Abstract
The p63 transcription factor is critical for epidermis formation in embryonic development, but its role in the adult epidermis is poorly understood. In this study, we show that acute genetic ablation of ΔNp63, the main p63 isoform, in adult epidermis disrupts keratinocyte proliferation and self-maintenance and, unexpectedly, triggers an inflammatory psoriasis-like condition. Mechanistically, single-cell RNA sequencing revealed the downregulation of cell cycle genes, upregulation of differentiation markers, and induction of several proinflammatory pathways in ΔNp63-ablated keratinocytes. Intriguingly, ΔNp63-ablated cells disappear by 3 weeks after ablation, at the expense of the remaining nonablated cells. This is not associated with active cell death and is likely due to reduced self-maintenance and enhanced differentiation. Indeed, in vivo wound healing, a physiological readout of the epidermal stem cell function, is severely impaired upon ΔNp63 ablation. We found that the Wnt signaling pathway (Wnt10A, Fzd6, Fzd10) and the activator protein 1 (JunB, Fos, FosB) factors are the likely ΔNp63 effectors responsible for keratinocyte proliferation/stemness and suppression of differentiation, respectively, whereas IL-1a, IL-18, IL-24, and IL-36γ are the likely negative effectors responsible for suppression of inflammation. These data establish ΔNp63 as a critical node that coordinates epidermal homeostasis, stemness, and suppression of inflammation, upstream of known regulatory pathways.
Collapse
Affiliation(s)
- Christopher E Eyermann
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA
| | - Xi Chen
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA
| | - Ozge S Somuncu
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA
| | - Jinyu Li
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA
| | | | - Jiang Chen
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA
| | - Evguenia M Alexandrova
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA.
| |
Collapse
|
19
|
Hélène C, Conrad O, Pflumio C, Borel C, Voegelin M, Bernard A, Schultz P, Onea MA, Jung A, Martin S, Burgy M. Dynamic profiling of immune microenvironment during anti-PD-1 immunotherapy for head and neck squamous cell carcinoma: the IPRICE study. BMC Cancer 2023; 23:1209. [PMID: 38066522 PMCID: PMC10704641 DOI: 10.1186/s12885-023-11672-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors of programmed cell death protein 1 (PD-1) represent a significant breakthrough in treating head and neck squamous cell carcinoma (HNSCC), with long-lasting responses and prolonged survival observed in first- and second-line therapy. However, this is observed in < 20% of patients and high primary/secondary resistance may occur. The primary objective of the identification of predictive factors for the response to anti-PD-1 immunotherapy in head and neck squamous cell carcinoma (IPRICE) study is to identify predictive factors of response to anti-PD-1 immunotherapy. METHODS The IPRICE study is a single-center, prospective, non-randomized, open-label, and interventional clinical trial. Liquid and tumor biopsies will be performed in 54 patients with recurrent/metastatic (R/M) HNSCC undergoing anti-PD-1 immunotherapy alone to compare the evolution of gene expression and immunological profile between responders and non-responders. We will use a multidisciplinary approach including spatial transcriptomics, single seq-RNA analysis, clinical data, and medical images. Genes, pathways, and transcription factors potentially involved in the immune response will also be analyzed, including genes involved in the interferon-gamma (IFN-γ) pathway, immunogenic cell death and mitophagy, hypoxia, circulating miRNA-mediated immunomodulation, cytokines, and immune repertoire within the tumor microenvironment (TME). With a follow-up period of 3-years, these data will help generate effective biomarkers to define optimal therapeutic strategy and new immunomodulatory agents based on a better understanding of primary/secondary resistance mechanisms. Tumor biopsy will be performed initially before the start of immunotherapy at the first tumor assessment and is only proposed at tumor progression. Clinical data will be collected using a dedicated Case Report Form (CRF). DISCUSSION Identifying predictive factors of the response to anti-PD-1 immunotherapy and optimizing long-term immune response require a thorough understanding of the intrinsic and acquired resistance to immunotherapy. To achieve this, dynamic profiling of TME during anti-PD-1 immunotherapy based on analysis of tumor biopsy samples is critical. This will be accomplished through the anatomical localization of HNSCC, which will allow for the analysis of multiple biopsies during treatment and the emergence of breakthrough technologies including single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics. TRIAL REGISTRATION Clinicaltrial.gov. Registered April 14, 2022, https://www. CLINICALTRIALS gov/study/NCT05328024 .
Collapse
Affiliation(s)
- Carinato Hélène
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe France, Strasbourg, France
| | - Ombline Conrad
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, Strasbourg, France
| | - Carole Pflumio
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe France, Strasbourg, France
| | - Christian Borel
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe France, Strasbourg, France
| | - Manon Voegelin
- Department of Clinical Research, Institut de Cancérologie Strasbourg Europe France, Strasbourg, France
| | - Alexandre Bernard
- Department of Clinical Research, Institut de Cancérologie Strasbourg Europe France, Strasbourg, France
| | - Philippe Schultz
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, Strasbourg, France
- Department of Otolaryngology and Cervico-Facial Surgery, Strasbourg University Hospital France, Strasbourg, France
| | - Mihaela-Alina Onea
- Department of Pathology, Strasbourg University Hospital France, Strasbourg, France
| | - Alain Jung
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, Strasbourg, France
- Laboratory of Tumor Biology, Institut de Cancérologie Strasbourg Europe, Strasbourg, 67200, France
| | - Sophie Martin
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, Strasbourg, France
| | - Mickaël Burgy
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe France, Strasbourg, France.
- Laboratory of Bioimaging and Pathology, University of Strasbourg, UMR7021 CNRS, Strasbourg, France.
| |
Collapse
|
20
|
Peng A, Lin X, Yang Q, Sun Y, Chen R, Liu B, Yu X. ΔNp63α facilitates proliferation and migration, and modulates the chromatin landscape in intrahepatic cholangiocarcinoma cells. Cell Death Dis 2023; 14:777. [PMID: 38012140 PMCID: PMC10682000 DOI: 10.1038/s41419-023-06309-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
p63 plays a crucial role in epithelia-originating tumours; however, its role in intrahepatic cholangiocarcinoma (iCCA) has not been completely explored. Our study revealed the oncogenic properties of p63 in iCCA and identified the major expressed isoform as ΔNp63α. We collected iCCA clinical data from The Cancer Genome Atlas database and analyzed p63 expression in iCCA tissue samples. We further established genetically modified iCCA cell lines in which p63 was overexpressed or knocked down to study the protein function/function of p63 in iCCA. We found that cells overexpressing p63, but not p63 knockdown counterparts, displayed increased proliferation, migration, and invasion. Transcriptome analysis showed that p63 altered the iCCA transcriptome, particularly by affecting cell adhesion-related genes. Moreover, chromatin accessibility decreased at p63 target sites when p63 binding was lost and increased when p63 binding was gained. The majority of the p63 bound sites were located in the distal intergenic regions and showed strong enhancer marks; however, active histone modifications around the Transcription Start Site changed as p63 expression changed. We also detected an interaction between p63 and the chromatin structural protein YY1. Taken together, our results suggest an oncogenic role for p63 in iCCA.
Collapse
Affiliation(s)
- Anghui Peng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Xiaowen Lin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Quanli Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Yihao Sun
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Ruiyan Chen
- Department of Dermatology, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Bing Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China.
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China.
| | - Xinyang Yu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China.
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China.
| |
Collapse
|
21
|
Guha A, Goswami KK, Sultana J, Ganguly N, Choudhury PR, Chakravarti M, Bhuniya A, Sarkar A, Bera S, Dhar S, Das J, Das T, Baral R, Bose A, Banerjee S. Cancer stem cell-immune cell crosstalk in breast tumor microenvironment: a determinant of therapeutic facet. Front Immunol 2023; 14:1245421. [PMID: 38090567 PMCID: PMC10711058 DOI: 10.3389/fimmu.2023.1245421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/25/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer (BC) is globally one of the leading killers among women. Within a breast tumor, a minor population of transformed cells accountable for drug resistance, survival, and metastasis is known as breast cancer stem cells (BCSCs). Several experimental lines of evidence have indicated that BCSCs influence the functionality of immune cells. They evade immune surveillance by altering the characteristics of immune cells and modulate the tumor landscape to an immune-suppressive type. They are proficient in switching from a quiescent phase (slowly cycling) to an actively proliferating phenotype with a high degree of plasticity. This review confers the relevance and impact of crosstalk between immune cells and BCSCs as a fate determinant for BC prognosis. It also focuses on current strategies for targeting these aberrant BCSCs that could open avenues for the treatment of breast carcinoma.
Collapse
Affiliation(s)
- Aishwarya Guha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | | | - Jasmine Sultana
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Nilanjan Ganguly
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Pritha Roy Choudhury
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Mohona Chakravarti
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Avishek Bhuniya
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anirban Sarkar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Saurav Bera
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Sukanya Dhar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Juhina Das
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Tapasi Das
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anamika Bose
- Department of Pharmaceutical Technology Biotechnology National Institute of Pharmaceutical Education and Research (NIPER) Sahibzada Ajit Singh (S.A.S.) Nagar, Mohali, Punjab, India
| | - Saptak Banerjee
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| |
Collapse
|
22
|
Li Y, Yang B, Miao H, Liu L, Wang Z, Jiang C, Yang Y, Qiu S, Li X, Geng Y, Zhang Y, Liu Y. Nicotinamide N -methyltransferase promotes M2 macrophage polarization by IL6 and MDSC conversion by GM-CSF in gallbladder carcinoma. Hepatology 2023; 78:1352-1367. [PMID: 36633260 DOI: 10.1097/hep.0000000000000028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/14/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND AIMS Nicotinamide N -methyltransferase (NNMT), an enzyme responsible for the methylation of nicotinamide, is involved in many metabolic pathways in adipose tissue and the liver. However, the role of NNMT in editing the tumor immune microenvironment is not well understood. APPROACH AND RESULTS Here, we identified that NNMT can promote IL6 and granulocyte-macrophage colony-stimulating factor (GM-CSF) expression by decreasing the tri-methyl-histone H3 levels on the promoters of IL6 and CSF2 (encoding GM-CSF) and CCAAT/Enhancer Binding Protein, an essential transcription factor for IL6 expression, thus promoting differentiation of macrophages into M2 type tumor-associated macrophages and generation of myeloid-derived suppressor cells from peripheral blood mononuclear cells. Treatment of xenografted tumor models overexpressing NNMT gallbladder carcinoma (GBC) cells with the NNMT inhibitor JBSNF-000088 resulted in compromised tumor development and decreased expression levels of IL6, GM-CSF, tumor-associated macrophage marker CD206, and myeloid-derived suppressor cell marker CD33 but increased expression levels of CD8. In addition, elevated expression of NNMT in tumors of patients with GBC was correlated with increased expression levels of CD206 and CD33 but with decreased levels of CD8 and survival of patients. CONCLUSIONS These data highlight the critical role of NNMT in GBC progression. Inhibition of NNMT by JBSNF-000088 is a potential molecular target for GBC immunotherapy.
Collapse
Affiliation(s)
- Yang Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
| | - Bo Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
| | - Huijie Miao
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liguo Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
| | - Ziyi Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengkai Jiang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shimei Qiu
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, China
| | - Xuechuan Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yajun Geng
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yijian Zhang
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, China
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| |
Collapse
|
23
|
Mourtada J, Lony C, Nicol A, De Azevedo J, Bour C, Macabre C, Roncarati P, Ledrappier S, Schultz P, Borel C, Burgy M, Wasylyk B, Mellitzer G, Herfs M, Gaiddon C, Jung AC. A novel ΔNp63-dependent immune mechanism improves prognosis of HPV-related head and neck cancer. Front Immunol 2023; 14:1264093. [PMID: 38022675 PMCID: PMC10630910 DOI: 10.3389/fimmu.2023.1264093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/29/2023] [Indexed: 12/01/2023] Open
Abstract
Background Deconvoluting the heterogenous prognosis of Human Papillomavirus (HPV)-related oropharyngeal squamous cell carcinoma (OSCC) is crucial for enhancing patient care, given its rapidly increasing incidence in western countries and the adverse side effects of OSCC treatments. Methods Transcriptomic data from HPV-positive OSCC samples were analyzed using unsupervised hierarchical clustering, and clinical relevance was evaluated using Kaplan-Meier analysis. HPV-positive OSCC cell line models were used in functional analyses and phenotypic assays to assess cell migration and invasion, response to cisplatin, and phagocytosis by macrophages in vitro. Results We found, by transcriptomic analysis of HPV-positive OSCC samples, a ΔNp63 dependent molecular signature that is associated with patient prognosis. ΔNp63 was found to act as a tumor suppressor in HPV-positive OSCC at multiple levels. It inhibits cell migration and invasion, and favors response to chemotherapy. RNA-Seq analysis uncovered an unexpected regulation of genes, such as DKK3, which are involved in immune response-signalling pathways. In agreement with these observations, we found that ΔNp63 expression levels correlate with an enhanced anti-tumor immune environment in OSCC, and ΔNp63 promotes cancer cell phagocytosis by macrophages through a DKK3/NF-κB-dependent pathway. Conclusion Our findings are the first comprehensive identification of molecular mechanisms involved in the heterogeneous prognosis of HPV-positive OSCC, paving the way for much-needed biomarkers and targeted treatment.
Collapse
Affiliation(s)
- Jana Mourtada
- Laboratoire de Biologie Tumorale, Institut de cancérologie Strasbourg Europe, Strasbourg, France
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, Laboratory « Streinth », Strasbourg, France
| | - Christelle Lony
- Laboratoire de Biologie Tumorale, Institut de cancérologie Strasbourg Europe, Strasbourg, France
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, Laboratory « Streinth », Strasbourg, France
| | - Anaïs Nicol
- Laboratoire de Radiobiologie, Institut de cancérologie Strasbourg Europe, Strasbourg, France
| | - Justine De Azevedo
- Laboratoire de Biologie Tumorale, Institut de cancérologie Strasbourg Europe, Strasbourg, France
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, Laboratory « Streinth », Strasbourg, France
| | - Cyril Bour
- Laboratoire de Biologie Tumorale, Institut de cancérologie Strasbourg Europe, Strasbourg, France
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, Laboratory « Streinth », Strasbourg, France
| | - Christine Macabre
- Laboratoire de Biologie Tumorale, Institut de cancérologie Strasbourg Europe, Strasbourg, France
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, Laboratory « Streinth », Strasbourg, France
- Tumorothèque du Centre Paul Strauss, Centre Paul Strauss, Strasbourg, France
| | - Patrick Roncarati
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Sonia Ledrappier
- Tumorothèque du Centre Paul Strauss, Centre Paul Strauss, Strasbourg, France
| | - Philippe Schultz
- Hôpitaux Universitaires de Strasbourg, Department of Otorhinolaryngology and Head and Neck Surgery, Strasbourg, France
| | - Christian Borel
- Department of Medical Oncology, Institut de cancérologie Strasbourg Europe, Strasbourg, France
| | - Mickaël Burgy
- Department of Medical Oncology, Institut de cancérologie Strasbourg Europe, Strasbourg, France
| | - Bohdan Wasylyk
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch-Graffenstaden, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U 1258, Illkirch-Graffenstaden, France
- Centre Nationale de la Recherche Scientifique (CNRS) UMR 7104, Illkirch-Graffenstaden, France
- Université de Strasbourg, Strasbourg, France
| | - Georg Mellitzer
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, Laboratory « Streinth », Strasbourg, France
| | - Michaël Herfs
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Christian Gaiddon
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, Laboratory « Streinth », Strasbourg, France
| | - Alain C. Jung
- Laboratoire de Biologie Tumorale, Institut de cancérologie Strasbourg Europe, Strasbourg, France
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, Laboratory « Streinth », Strasbourg, France
- Tumorothèque du Centre Paul Strauss, Centre Paul Strauss, Strasbourg, France
| |
Collapse
|
24
|
Ji S, Yu H, Zhou D, Fan X, Duan Y, Tan Y, Lang M, Shao G. Cancer stem cell-derived CHI3L1 activates the MAF/CTLA4 signaling pathway to promote immune escape in triple-negative breast cancer. J Transl Med 2023; 21:721. [PMID: 37838657 PMCID: PMC10576881 DOI: 10.1186/s12967-023-04532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 09/17/2023] [Indexed: 10/16/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) development may be associated with tumor immune escape. This study explores whether the CHI3L1/MAF/CTLA4/S100A4 axis affects immune escape in TNBC through interplay with triple-negative breast cancer stem cells (TN-BCSCs). OBJECTIVE The aim of this study is to utilize single-cell transcriptome sequencing (scRNA-seq) to uncover the molecular mechanisms by which the CHI3L1/MAF/CTLA4 signaling pathway may mediate immune evasion in triple-negative breast cancer through the interaction between tumor stem cells (CSCs) and immune cells. METHODS Cell subsets in TNBC tissues were obtained through scRNA-seq, followed by screening differentially expressed genes in TN-BCSCs and B.C.s (CD44+ and CD24-) and predicting the transcription factor regulated by CHI3L1. Effect of CHI3L1 on the stemness phenotype of TNBC cells investigated. Effects of BCSCs-231-derived CHI3L1 on CTLA4 expression in T cells were explored after co-culture of BCSCs-231 cells obtained from microsphere culture of TN-BCSCs with T cells. BCSCs-231-treated T cells were co-cultured with CD8+ T cells to explore the resultant effect on T cell cytotoxicity. An orthotopic B.C. transplanted tumor model in mice with humanized immune systems was constructed, in which the Role of CHI3L1/MAF/CTLA4 in the immune escape of TNBC was explored. RESULTS Eight cell subsets were found in the TNBC tissues, and the existence of TN-BCSCs was observed in the epithelial cell subset. CHI3L1 was related to the stemness phenotype of TNBC cells. TN-BCSC-derived CHI3L1 increased CTLA4 expression in T cells through MAF, inhibiting CD8+ T cell cytotoxicity and inducing immunosuppression. Furthermore, the CTLA4+ T cells might secrete S100A4 to promote the stemness phenotype of TNBC cells. CONCLUSIONS TN-BCSC-derived CHI3L1 upregulates CTLA4 expression in T cells through MAF, suppressing the function of CD8+ T cells, which promotes the immune escape of TNBC.
Collapse
Affiliation(s)
- Shufeng Ji
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China
| | - Hao Yu
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China
| | - Dan Zhou
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Xulong Fan
- Department of Breast Surgery, Maternity and Children's Healthcare Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Yan Duan
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China
| | - Yijiang Tan
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China
| | - Min Lang
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China
| | - Guoli Shao
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China.
| |
Collapse
|
25
|
Sakakibara N, Clavijo PE, Sievers C, Gray VC, King KE, George AL, Ponnamperuma RM, Walter BA, Chen Z, Van Waes C, Allen CT, Weinberg WC. Oncogenic Ras and ΔNp63α cooperate to recruit immunosuppressive polymorphonuclear myeloid-derived suppressor cells in a mouse model of squamous cancer pathogenesis. Front Immunol 2023; 14:1200970. [PMID: 37638000 PMCID: PMC10449460 DOI: 10.3389/fimmu.2023.1200970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/13/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Amplification of human chromosome 3q26-29, which encodes oncoprotein ΔNp63 among other isoforms of the p63 family, is a feature common to squamous cell carcinomas (SCCs) of multiple tissue origins. Along with overexpression of ΔNp63, activation of the protooncogene, RAS, whether by overexpression or oncogenic mutation, is frequently observed in many cancers. In this study, analysis of transcriptome data from The Cancer Genome Atlas (TCGA) demonstrated that expression of TP63 mRNA, particularly ΔNp63 isoforms, and HRAS are significantly elevated in advanced squamous cell carcinomas of the head and neck (HNSCCs), suggesting pathological significance. However, how co-overexpressed ΔNp63 and HRAS affect the immunosuppressive tumor microenvironment (TME) is incompletely understood. Methods Here, we established and characterized an immune competent mouse model using primary keratinocytes with retroviral-mediated overexpression of ΔNp63α and constitutively activated HRAS (v-rasHa G12R) to evaluate the role of these oncogenes in the immune TME. Results In this model, orthotopic grafting of wildtype syngeneic keratinocytes expressing both v-rasHa and elevated levels of ΔNp63α consistently yield carcinomas in syngeneic hosts, while cells expressing v-rasHa alone yield predominantly papillomas. We found that polymorphonuclear (PMN) myeloid cells, experimentally validated to be immunosuppressive and thus representing myeloid-derived suppressor cells (PMN-MDSCs), were significantly recruited into the TME of carcinomas arising early following orthotopic grafting of ΔNp63α/v-rasHa-expressing keratinocytes. ΔNp63α/v-rasHa-driven carcinomas expressed higher levels of chemokines implicated in recruitment of MDSCs compared to v-rasHa-initiated tumors, providing a heretofore undescribed link between ΔNp63α/HRAS-driven carcinomas and the development of an immunosuppressive TME. Conclusion These results support the utilization of a genetic carcinogenesis model harboring specific genomic drivers of malignancy to study mechanisms underlying the development of local immunosuppression.
Collapse
Affiliation(s)
- Nozomi Sakakibara
- Office of Biotechnology Products, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, United States
| | - Paúl E. Clavijo
- Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, United States
| | - Cem Sievers
- Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, United States
| | - Veronica C. Gray
- Office of Biotechnology Products, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, United States
| | - Kathryn E. King
- Office of Biotechnology Products, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, United States
| | - Andrea L. George
- Office of Biotechnology Products, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, United States
| | - Roshini M. Ponnamperuma
- Office of Biotechnology Products, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, United States
| | - Beatriz A. Walter
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, MD, United States
| | - Zhong Chen
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, United States
| | - Carter Van Waes
- Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, United States
| | - Clint T. Allen
- Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, United States
| | - Wendy C. Weinberg
- Office of Biotechnology Products, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, United States
| |
Collapse
|
26
|
Githaka JM, Pirayeshfard L, Goping IS. Cancer invasion and metastasis: Insights from murine pubertal mammary gland morphogenesis. Biochim Biophys Acta Gen Subj 2023; 1867:130375. [PMID: 37150225 DOI: 10.1016/j.bbagen.2023.130375] [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: 12/20/2022] [Revised: 04/20/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Cancer invasion and metastasis accounts for the majority of cancer related mortality. A better understanding of the players that drive the aberrant invasion and migration of tumors cells will provide critical targets to inhibit metastasis. Postnatal pubertal mammary gland morphogenesis is characterized by highly proliferative, invasive, and migratory normal epithelial cells. Identifying the molecular regulators of pubertal gland development is a promising strategy since tumorigenesis and metastasis is postulated to be a consequence of aberrant reactivation of developmental stages. In this review, we summarize the pubertal morphogenesis regulators that are involved in cancer metastasis and revisit pubertal mammary gland transcriptome profiling to uncover both known and unknown metastasis genes. Our updated list of pubertal morphogenesis regulators shows that most are implicated in invasion and metastasis. This review highlights molecular linkages between development and metastasis and provides a guide for exploring novel metastatic drivers.
Collapse
Affiliation(s)
- John Maringa Githaka
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Leila Pirayeshfard
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ing Swie Goping
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Department of Oncology, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
27
|
Liu H, Wang Z, Zhou Y, Yang Y. MDSCs in breast cancer: an important enabler of tumor progression and an emerging therapeutic target. Front Immunol 2023; 14:1199273. [PMID: 37465670 PMCID: PMC10350567 DOI: 10.3389/fimmu.2023.1199273] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/19/2023] [Indexed: 07/20/2023] Open
Abstract
Women worldwide are more likely to develop breast cancer (BC) than any other type of cancer. The treatment of BC depends on the subtype and stage of the cancer, such as surgery, radiotherapy, chemotherapy, and immunotherapy. Although significant progress has been made in recent years, advanced or metastatic BC presents a poor prognosis, due to drug resistance and recurrences. During embryonic development, myeloid-derived suppressor cells (MDSCs) develop that suppress the immune system. By inhibiting anti-immune effects and promoting non-immune mechanisms such as tumor cell stemness, epithelial-mesenchymal transformation (EMT) and angiogenesis, MDSCs effectively promote tumor growth and metastasis. In various BC models, peripheral tissues, and tumor microenvironments (TME), MDSCs have been found to amplification. Clinical progression or poor prognosis are strongly associated with increased MDSCs. In this review, we describe the activation, recruitment, and differentiation of MDSCs production in BC, the involvement of MDSCs in BC progression, and the clinical characteristics of MDSCs as a potential BC therapy target.
Collapse
Affiliation(s)
- Haoyu Liu
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, China
| | - Zhicheng Wang
- National Health Commission (NHC) Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Yuntao Zhou
- National Health Commission (NHC) Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Yanming Yang
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
28
|
Tacchini M, Sacchetti G, Guerrini A, Paganetto G. Mycochemicals against Cancer Stem Cells. Toxins (Basel) 2023; 15:360. [PMID: 37368660 DOI: 10.3390/toxins15060360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/08/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Since ancient times, mushrooms have been considered valuable allies of human well-being both from a dietary and medicinal point of view. Their essential role in several traditional medicines is explained today by the discovery of the plethora of biomolecules that have shown proven efficacy for treating various diseases, including cancer. Numerous studies have already been conducted to explore the antitumoural properties of mushroom extracts against cancer. Still, very few have reported the anticancer properties of mushroom polysaccharides and mycochemicals against the specific population of cancer stem cells (CSCs). In this context, β-glucans are relevant in modulating immunological surveillance against this subpopulation of cancer cells within tumours. Small molecules, less studied despite their spread and assortment, could exhibit the same importance. In this review, we discuss several pieces of evidence of the association between β-glucans and small mycochemicals in modulating biological mechanisms which are proven to be involved with CSCs development. Experimental evidence and an in silico approach are evaluated with the hope of contributing to future strategies aimed at the direct study of the action of these mycochemicals on this subpopulation of cancer cells.
Collapse
Affiliation(s)
- Massimo Tacchini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Gianni Sacchetti
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Alessandra Guerrini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Guglielmo Paganetto
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
29
|
Fisher ML, Balinth S, Mills AA. ΔNp63α in cancer: importance and therapeutic opportunities. Trends Cell Biol 2023; 33:280-292. [PMID: 36115734 PMCID: PMC10011024 DOI: 10.1016/j.tcb.2022.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/09/2022] [Accepted: 08/22/2022] [Indexed: 10/14/2022]
Abstract
Our understanding of cancer and the key pathways that drive cancer survival has expanded rapidly over the past several decades. However, there are still important challenges that continue to impair patient survival, including our inability to target cancer stem cells (CSCs), metastasis, and drug resistance. The transcription factor p63 is a p53 family member with multiple isoforms that carry out a wide array of functions. Here, we discuss the critical importance of the ΔNp63α isoform in cancer and potential therapeutic strategies to target ΔNp63α expression to impair the CSC population, as well as to prevent metastasis and drug resistance to improve patient survival.
Collapse
Affiliation(s)
- Matthew L Fisher
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Seamus Balinth
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, NY 11794, USA
| | - Alea A Mills
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
30
|
Thacker G, Henry S, Nandi A, Debnath R, Singh S, Nayak A, Susnik B, Boone MM, Zhang Q, Kesmodel SB, Gumber S, Das GM, Kambayashi T, Dos Santos CO, Chakrabarti R. Immature natural killer cells promote progression of triple-negative breast cancer. Sci Transl Med 2023; 15:eabl4414. [PMID: 36888695 PMCID: PMC10875969 DOI: 10.1126/scitranslmed.abl4414] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/26/2023] [Indexed: 03/10/2023]
Abstract
Natural killer (NK) cells are cytotoxic lymphocytes that accumulate within the tumor microenvironment and are generally considered to be antitumorigenic. Using single-cell RNA sequencing and functional analysis of multiple triple-negative breast cancer (TNBC) and basal tumor samples, we observed a unique subcluster of Socs3highCD11b-CD27- immature NK cells that were present only in TNBC samples. These tumor-infiltrating NK cells expressed a reduced cytotoxic granzyme signature and, in mice, were responsible for activating cancer stem cells through Wnt signaling. NK cell-mediated activation of these cancer stem cells subsequently enhanced tumor progression in mice, whereas depletion of NK cells or Wnt ligand secretion from NK cells by LGK-974 decreased tumor progression. In addition, NK cell depletion or inhibition of their function improved anti-programmed cell death ligand 1 (PD-L1) antibody or chemotherapy response in mice with TNBC. Furthermore, tumor samples from patients with TNBC and non-TNBC revealed that increased numbers of CD56bright NK cells were present in TNBC tumors and were correlated to poor overall survival in patients with TNBC. Together, our findings identify a population of protumorigenic NK cells that may be exploited for both diagnostic and therapeutic strategies to improve outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Gatha Thacker
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Samantha Henry
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Ajeya Nandi
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rahul Debnath
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Snahlata Singh
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anupma Nayak
- Department of Pathology and Laboratory Medicine at the Hospital of the University of Pennsylvania, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Barbara Susnik
- Department of Pathology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Melinda M Boone
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Qing Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Susan B Kesmodel
- DeWitt Daughtry Family Department of Surgery, Division of Surgical Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Sanjeev Gumber
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Gokul M Das
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Camila O. Dos Santos
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Rumela Chakrabarti
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
31
|
Segal BH, Giridharan T, Suzuki S, Khan ANH, Zsiros E, Emmons TR, Yaffe MB, Gankema AAF, Hoogeboom M, Goetschalckx I, Matlung HL, Kuijpers TW. Neutrophil interactions with T cells, platelets, endothelial cells, and of course tumor cells. Immunol Rev 2023; 314:13-35. [PMID: 36527200 PMCID: PMC10174640 DOI: 10.1111/imr.13178] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neutrophils sense microbes and host inflammatory mediators, and traffic to sites of infection where they direct a broad armamentarium of antimicrobial products against pathogens. Neutrophils are also activated by damage-associated molecular patterns (DAMPs), which are products of cellular injury that stimulate the innate immune system through pathways that are similar to those activated by microbes. Neutrophils and platelets become activated by injury, and cluster and cross-signal to each other with the cumulative effect of driving antimicrobial defense and hemostasis. In addition, neutrophil extracellular traps are extracellular chromatin and granular constituents that are generated in response to microbial and damage motifs and are pro-thrombotic and injurious. Although neutrophils can worsen tissue injury, neutrophils may also have a role in facilitating wound repair following injury. A central theme of this review relates to how critical functions of neutrophils that evolved to respond to infection and damage modulate the tumor microenvironment (TME) in ways that can promote or limit tumor progression. Neutrophils are reprogrammed by the TME, and, in turn, can cross-signal to tumor cells and reshape the immune landscape of tumors. Importantly, promising new therapeutic strategies have been developed to target neutrophil recruitment and function to make cancer immunotherapy more effective.
Collapse
Affiliation(s)
- Brahm H Segal
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Thejaswini Giridharan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Sora Suzuki
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Anm Nazmul H Khan
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Tiffany R Emmons
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Michael B Yaffe
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Angela A F Gankema
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark Hoogeboom
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Ines Goetschalckx
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanke L Matlung
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Emma Children's Hospital Amsterdam University Medical Center (Amsterdam UMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
32
|
PMN-MDSCs modulated by CCL20 from cancer cells promoted breast cancer cell stemness through CXCL2-CXCR2 pathway. Signal Transduct Target Ther 2023; 8:97. [PMID: 36859354 PMCID: PMC9977784 DOI: 10.1038/s41392-023-01337-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 01/05/2023] [Accepted: 01/29/2023] [Indexed: 03/03/2023] Open
Abstract
Our previous studies have showed that C-C motif chemokine ligand 20 (CCL20) advanced tumor progression and enhanced the chemoresistance of cancer cells by positively regulating breast cancer stem cell (BCSC) self-renewal. However, it is unclear whether CCL20 affects breast cancer progression by remodeling the tumor microenvironment (TME). Here, we observed that polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were remarkably enriched in TME of CCL20-overexpressing cancer cell orthotopic allograft tumors. Mechanistically, CCL20 activated the differentiation of granulocyte-monocyte progenitors (GMPs) via its receptor C-C motif chemokine receptor 6 (CCR6) leading to the PMN-MDSC expansion. PMN-MDSCs from CCL20-overexpressing cell orthotopic allograft tumors (CCL20-modulated PMN-MDSCs) secreted amounts of C-X-C motif chemokine ligand 2 (CXCL2) and increased ALDH+ BCSCs via activating CXCR2/NOTCH1/HEY1 signaling pathway. Furthermore, C-X-C motif chemokine receptor 2 (CXCR2) antagonist SB225002 enhanced the docetaxel (DTX) effects on tumor growth by decreasing BCSCs in CCL20high-expressing tumors. These findings elucidated how CCL20 modulated the TME to promote cancer development, indicating a new therapeutic strategy by interfering with the interaction between PMN-MDSCs and BCSCs in breast cancer, especially in CCL20high-expressing breast cancer.
Collapse
|
33
|
Han J, Wan M, Ma Z, Yi H. Regulation of DNA-PK activity promotes the progression of TNBC via enhancing the immunosuppressive function of myeloid-derived suppressor cells. Cancer Med 2023; 12:5939-5952. [PMID: 36373232 PMCID: PMC10028116 DOI: 10.1002/cam4.5387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 10/02/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND DNA-dependent protein kinase (DNA-PK) is engaged in DNA damage repair and is significantly expressed in triple negative breast cancer (TNBC). Inhibiting DNA-PK to reduce DNA damage repair provides a possibility of tumor treatment. NU7441, a DNA-PK inhibitor, can regulate the function and differentiation of CD4+ T cells and effectively enhance immunogenicity of monocyte-derived dendritic cells. However, the effect of NU7441 on the tumor progression activity of immunosuppressive myeloid-derived suppressor cells (MDSCs) in TNBC remains unclear. RESULTS In this study, we found that NU7441 alone significantly increased tumor growth in 4 T1 (a mouse TNBC cell line) tumor-bearing mice. Bioinformatics analysis showed that DNA-PK and functional markers of MDSCs (iNOS, Arg1, and IDO) tended to coexist in breast cancer patients. The mutations of these genes were significantly correlated with lower survival in breast cancer patients. Moreover, NU7441 significantly decreased the percentage of MDSCs in peripheral blood mononuclear cells (PBMCs), spleen and tumor, but enhanced the immunosuppressive function of splenic MDSCs. Furthermore, NU7441 increased MDSCs' DNA-PK and pDNA-PK protein levels in PBMCs and in the spleen and increased DNA-PK mRNA expression and expression of MDSCs functional markers in splenic MDSCs from tumor-bearing mice. NU7441 combined with gemcitabine reduced tumor volume, which may be because gemcitabine eliminated the remaining MDSCs with enhanced immunosuppressive ability. CONCLUSIONS These findings highlight that the regulation of DNA-PK activity by NU7441 promotes TNBC progression via enhancing the immunosuppressive function of MDSCs. Moreover, NU7441 combined with gemcitabine offers an efficient therapeutic approach for TNBC and merits deeper investigation.
Collapse
Affiliation(s)
- Jiawen Han
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration and Transplantation Ministry of Education, Changchun, China
| | - Minjie Wan
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Zhanchuan Ma
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration and Transplantation Ministry of Education, Changchun, China
| | - Huanfa Yi
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration and Transplantation Ministry of Education, Changchun, China
| |
Collapse
|
34
|
Glathar AR, Oyelakin A, Nayak KB, Sosa J, Romano RA, Sinha S. A Systemic and Integrated Analysis of p63-Driven Regulatory Networks in Mouse Oral Squamous Cell Carcinoma. Cancers (Basel) 2023; 15:446. [PMID: 36672394 PMCID: PMC9856320 DOI: 10.3390/cancers15020446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/04/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignancy of the oral cavity and is linked to tobacco exposure, alcohol consumption, and human papillomavirus infection. Despite therapeutic advances, a lack of molecular understanding of disease etiology, and delayed diagnoses continue to negatively affect survival. The identification of oncogenic drivers and prognostic biomarkers by leveraging bulk and single-cell RNA-sequencing datasets of OSCC can lead to more targeted therapies and improved patient outcomes. However, the generation, analysis, and continued utilization of additional genetic and genomic tools are warranted. Tobacco-induced OSCC can be modeled in mice via 4-nitroquinoline 1-oxide (4NQO), which generates a spectrum of neoplastic lesions mimicking human OSCC and upregulates the oncogenic master transcription factor p63. Here, we molecularly characterized established mouse 4NQO treatment-derived OSCC cell lines and utilized RNA and chromatin immunoprecipitation-sequencing to uncover the global p63 gene regulatory and signaling network. We integrated our p63 datasets with published bulk and single-cell RNA-sequencing of mouse 4NQO-treated tongue and esophageal tumors, respectively, to generate a p63-driven gene signature that sheds new light on the role of p63 in murine OSCC. Our analyses reveal known and novel players, such as COTL1, that are regulated by p63 and influence various oncogenic processes, including metastasis. The identification of new sets of potential biomarkers and pathways, some of which are functionally conserved in human OSCC and can prognosticate patient survival, offers new avenues for future mechanistic studies.
Collapse
Affiliation(s)
- Alexandra Ruth Glathar
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Akinsola Oyelakin
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Kasturi Bala Nayak
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Jennifer Sosa
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Rose-Anne Romano
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
35
|
Onkar SS, Carleton NM, Lucas PC, Bruno TC, Lee AV, Vignali DAA, Oesterreich S. The Great Immune Escape: Understanding the Divergent Immune Response in Breast Cancer Subtypes. Cancer Discov 2023; 13:23-40. [PMID: 36620880 PMCID: PMC9833841 DOI: 10.1158/2159-8290.cd-22-0475] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/30/2022] [Accepted: 09/26/2022] [Indexed: 12/12/2022]
Abstract
Breast cancer, the most common type of cancer affecting women, encompasses a collection of histologic (mainly ductal and lobular) and molecular subtypes exhibiting diverse clinical presentation, disease trajectories, treatment options, and outcomes. Immunotherapy has revolutionized treatment for some solid tumors but has shown limited promise for breast cancers. In this review, we summarize recent advances in our understanding of the complex interactions between tumor and immune cells in subtypes of breast cancer at the cellular and microenvironmental levels. We aim to provide a perspective on opportunities for future immunotherapy agents tailored to specific features of each subtype of breast cancer. SIGNIFICANCE Although there are currently over 200 ongoing clinical trials testing immunotherapeutics, such as immune-checkpoint blockade agents, these are largely restricted to the triple-negative and HER2+ subtypes and primarily focus on T cells. With the rapid expansion of new in vitro, in vivo, and clinical data, it is critical to identify and highlight the challenges and opportunities unique for each breast cancer subtype to drive the next generation of treatments that harness the immune system.
Collapse
Affiliation(s)
- Sayali S. Onkar
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Neil M. Carleton
- Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Peter C Lucas
- Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Adrian V Lee
- Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Dario AA Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Steffi Oesterreich
- Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
36
|
Zhu S, Lalani AI, Jin J, Sant’Angelo D, Covey LR, Liu K, Young HA, Ostrand-Rosenberg S, Xie P. The adaptor protein TRAF3 is an immune checkpoint that inhibits myeloid-derived suppressor cell expansion. Front Immunol 2023; 14:1167924. [PMID: 37207205 PMCID: PMC10189059 DOI: 10.3389/fimmu.2023.1167924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are aberrantly expanded in cancer patients and under other pathological conditions. These cells orchestrate the immunosuppressive and inflammatory network to facilitate cancer metastasis and mediate patient resistance to therapies, and thus are recognized as a prime therapeutic target of human cancers. Here we report the identification of the adaptor protein TRAF3 as a novel immune checkpoint that critically restrains MDSC expansion. We found that myeloid cell-specific Traf3-deficient (M-Traf3 -/-) mice exhibited MDSC hyperexpansion during chronic inflammation. Interestingly, MDSC hyperexpansion in M-Traf3 -/- mice led to accelerated growth and metastasis of transplanted tumors associated with an altered phenotype of T cells and NK cells. Using mixed bone marrow chimeras, we demonstrated that TRAF3 inhibited MDSC expansion via both cell-intrinsic and cell-extrinsic mechanisms. Furthermore, we elucidated a GM-CSF-STAT3-TRAF3-PTP1B signaling axis in MDSCs and a novel TLR4-TRAF3-CCL22-CCR4-G-CSF axis acting in inflammatory macrophages and monocytes that coordinately control MDSC expansion during chronic inflammation. Taken together, our findings provide novel insights into the complex regulatory mechanisms of MDSC expansion and open up unique perspectives for the design of new therapeutic strategies that aim to target MDSCs in cancer patients.
Collapse
Affiliation(s)
- Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Almin I. Lalani
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Juan Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Pharmacology, Anhui Medical University, Hefei, Anhui, China
| | - Derek Sant’Angelo
- Child Health Institute of New Jersey, Rutgers University, New Brunswick, NJ, United States
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, United States
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Lori R. Covey
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, United States
| | - Howard A. Young
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD, United States
| | - Suzanne Ostrand-Rosenberg
- Department of Biological Sciences, The University of Maryland, Baltimore County, Baltimore, MD, United States
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
- *Correspondence: Ping Xie,
| |
Collapse
|
37
|
Li Y, Li M, Su K, Zong S, Zhang H, Xiong L. Pre-metastatic niche: from revealing the molecular and cellular mechanisms to the clinical applications in breast cancer metastasis. Theranostics 2023; 13:2301-2318. [PMID: 37153744 PMCID: PMC10157731 DOI: 10.7150/thno.82700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/08/2023] [Indexed: 05/10/2023] Open
Abstract
Breast cancer (BC) is one of the most commonly diagnosed cancers and the leading cause of cancer-related deaths in women worldwide. Metastasis is a major contributor to high cancer mortality and is usually the endpoint of a series of sequential and dynamic events. One of the critical events is forming a pre-metastatic niche (PMN) that occurs before macroscopic tumor cell invasion and provides a suitable environment for tumor cells to colonize and progress into metastases. Due to the unique characteristics of PMN in cancer metastasis, developing therapies to target PMN may bring new advantages in preventing cancer metastasis at an early stage. Various biological molecules, cells, and signaling pathways are altered in BC, regulating the functions of distinctive immune cells and stromal remodeling, inducing angiogenesis, and effect metabolic reprogramming and organotropism to promote PMN formation. In this review, we elucidate the multifaceted mechanisms contributing to the development of PMN in BC, discuss the characteristics of PMN, and highlight the significance of PMN in providing potential diagnostic and therapeutic strategies for BC metastasis, which may bring promising insights and foundations for future studies.
Collapse
Affiliation(s)
- Yuqiu Li
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
- Queen Mary College of Nanchang University, Nanchang 330006, China
| | - Miao Li
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Kangtai Su
- First Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Siwen Zong
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Hongyan Zhang
- Department of Burn, The First Affiliated Hospital, Nanchang University, 17 Yongwaizheng Road, Nanschang 330066, China
- ✉ Corresponding authors: Hongyan Zhang and Lixia Xiong; and
| | - Lixia Xiong
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
- ✉ Corresponding authors: Hongyan Zhang and Lixia Xiong; and
| |
Collapse
|
38
|
Ma W, Han X, Shasaltaneh MD, Hosseinifard H, Maghsoudloo M, Zhang Y, Weng Q, Wang Q, Wen Q, Imani S. The p110α/ΔNp63α complex mutations in triple-negative breast cancer: Potential targets for transcriptional-based therapies. Tumour Biol 2023; 45:127-146. [PMID: 37980588 DOI: 10.3233/tub-230013] [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: 11/21/2023] Open
Abstract
BACKGROUND Hotspot mutations occurring in the p110α domain of the PIK3CA gene, specifically p110αH1047R/L increase tumor metastasis and cell motility in triple-negative breast cancer (TNBC). These mutations also affect the transcriptional regulation of ΔNp63α, a significant isoform of the p53 protein involved in cancer progression. This study attempts to investigate the transcriptional impact of p110αH1047R/L mutations on the PIK3CA/ΔNp63α complex in TNBC carcinogenesis. METHODS We performed site-directed mutagenesis to introduce p110αH1047R/L mutations and evaluated their oncogenic effects on the growth, invasion, migration, and apoptosis of three different TNBC cell lines in vitro. We investigated the impact of these mutations on the p110α/ΔNp63α complex and downstream transcriptional signaling pathways at the gene and protein levels. Additionally, we used bioinformatics techniques such as molecular dynamics simulations and protein-protein docking to gain insight into the stability and structural changes induced by the p110αH1047R/L mutations in the p110α/ΔNp63α complex and downstream signaling pathway. RESULTS The presence of PIK3CA oncogenic hotspot mutations in the p110α/ΔNp63α complex led to increased scattering of TNBC cells during growth, migration, and invasion. Our in vitro mutagenesis assay showed that the p110αH1047R/L mutations activated the PI3K-Akt-mTOR and tyrosine kinase receptor pathways, resulting in increased cell proliferation, invasion, and apoptosis in TNBC cells. These mutations decreased the repressing effect of ΔNp63α on the p110α kinase domain, leading to the enhancement of downstream signaling pathways of PI3K and tyrosine kinase receptors and oncogenic transformation in TNBC. Additionally, our findings suggest that the physical interaction between the DNA binding domain of ΔNp63α and the kinase domain of p110α may be partially impaired, potentially leading to alterations in the conformation of the p110α/ΔNp63α complex. CONCLUSION Our findings suggest that targeting the p110αH1047R/L mutations in TNBC could be a promising strategy for developing transcriptional-based therapies. Restoring the interaction between ΔNp63α and the p110α kinase domain, which is disrupted by these mutations, may provide a new approach to treating TNBC.
Collapse
Affiliation(s)
- Wenqiong Ma
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xingping Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | | | - Hossein Hosseinifard
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuqin Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiao Weng
- Department of Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qingjing Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - QingLian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|
39
|
Zhang L, Chen W, Liu S, Chen C. Targeting Breast Cancer Stem Cells. Int J Biol Sci 2023; 19:552-570. [PMID: 36632469 PMCID: PMC9830502 DOI: 10.7150/ijbs.76187] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 12/09/2022] [Indexed: 01/04/2023] Open
Abstract
The potential roles of breast cancer stem cells (BCSCs) in tumor initiation and recurrence have been recognized for many decades. Due to their strong capacity for self-renewal and differentiation, BCSCs are the major reasons for poor clinical outcomes and low therapeutic response. Several hypotheses on the origin of cancer stem cells have been proposed, including critical gene mutations in stem cells, dedifferentiation of somatic cells, and cell plasticity remodeling by epithelial-mesenchymal transition (EMT) and the tumor microenvironment. Moreover, the tumor microenvironment, including cellular components and cytokines, modulates the self-renewal and therapeutic resistance of BCSCs. Small molecules, antibodies, and chimeric antigen receptor (CAR)-T cells targeting BCSCs have been developed, and their applications in combination with conventional therapies are undergoing clinical trials. In this review, we focus on the features of BCSCs, emphasize the major factors and tumor environment that regulate the stemness of BCSCs, and discuss potential BCSC-targeting therapies.
Collapse
Affiliation(s)
- Lu Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai paracrine Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai 200032, China
| | - Wenmin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming 650201, China.,Kunming College of Life Sciences, the University of the Chinese Academy of Sciences, Kunming 650201, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai paracrine Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai 200032, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China.,✉ Corresponding authors: Ceshi Chen, E-mail: or Suling Liu, E-mail:
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming 650201, China.,Academy of Biomedical Engineering, Kunming Medical University, Kunming 650500, China.,The Third Affiliated Hospital, Kunming Medical University, Kunming 650118, China.,✉ Corresponding authors: Ceshi Chen, E-mail: or Suling Liu, E-mail:
| |
Collapse
|
40
|
Mechanisms and Strategies to Overcome PD-1/PD-L1 Blockade Resistance in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 15:cancers15010104. [PMID: 36612100 PMCID: PMC9817764 DOI: 10.3390/cancers15010104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by a high rate of systemic metastasis, insensitivity to conventional treatment and susceptibility to drug resistance, resulting in a poor patient prognosis. The immune checkpoint inhibitors (ICIs) represented by antibodies of programmed death receptor 1 (PD-1) and programmed death receptor ligand 1 (PD-L1) have provided new therapeutic options for TNBC. However, the efficacy of PD-1/PD-L1 blockade monotherapy is suboptimal immune response, which may be caused by reduced antigen presentation, immunosuppressive tumor microenvironment, interplay with other immune checkpoints and aberrant activation of oncological signaling in tumor cells. Therefore, to improve the sensitivity of TNBC to ICIs, suitable patients are selected based on reliable predictive markers and treated with a combination of ICIs with other therapies such as chemotherapy, radiotherapy, targeted therapy, oncologic virus and neoantigen-based therapies. This review discusses the current mechanisms underlying the resistance of TNBC to PD-1/PD-L1 inhibitors, the potential biomarkers for predicting the efficacy of anti-PD-1/PD-L1 immunotherapy and recent advances in the combination therapies to increase response rates, the depth of remission and the durability of the benefit of TNBC to ICIs.
Collapse
|
41
|
Lambert AW, Fiore C, Chutake Y, Verhaar ER, Strasser PC, Chen MW, Farouq D, Das S, Li X, Eaton EN, Zhang Y, Liu Donaher J, Engstrom I, Reinhardt F, Yuan B, Gupta S, Wollison B, Eaton M, Bierie B, Carulli J, Olson ER, Guenther MG, Weinberg RA. ΔNp63/p73 drive metastatic colonization by controlling a regenerative epithelial stem cell program in quasi-mesenchymal cancer stem cells. Dev Cell 2022; 57:2714-2730.e8. [PMID: 36538894 PMCID: PMC10002472 DOI: 10.1016/j.devcel.2022.11.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/03/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022]
Abstract
Cancer stem cells (CSCs) may serve as the cellular seeds of tumor recurrence and metastasis, and they can be generated via epithelial-mesenchymal transitions (EMTs). Isolating pure populations of CSCs is difficult because EMT programs generate multiple alternative cell states, and phenotypic plasticity permits frequent interconversions between these states. Here, we used cell-surface expression of integrin β4 (ITGB4) to isolate highly enriched populations of human breast CSCs, and we identified the gene regulatory network operating in ITGB4+ CSCs. Specifically, we identified ΔNp63 and p73, the latter of which transactivates ΔNp63, as centrally important transcriptional regulators of quasi-mesenchymal CSCs that reside in an intermediate EMT state. We found that the transcriptional program controlled by ΔNp63 in CSCs is largely distinct from the one that it orchestrates in normal basal mammary stem cells and, instead, it more closely resembles a regenerative epithelial stem cell response to wounding. Moreover, quasi-mesenchymal CSCs repurpose this program to drive metastatic colonization via autocrine EGFR signaling.
Collapse
Affiliation(s)
- Arthur W Lambert
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | | | | | - Elisha R Verhaar
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | | | | | | | - Sunny Das
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Xin Li
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Elinor Ng Eaton
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Yun Zhang
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Joana Liu Donaher
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Ian Engstrom
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Ferenc Reinhardt
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Bingbing Yuan
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Sumeet Gupta
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | | | | | - Brian Bierie
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | | | | | | | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; MIT Ludwig Center for Molecular Oncology, Cambridge, MA 02139, USA.
| |
Collapse
|
42
|
Zhao J, Dong Y, Zhang Y, Wang J, Wang Z. Biophysical heterogeneity of myeloid-derived microenvironment to regulate resistance to cancer immunotherapy. Adv Drug Deliv Rev 2022; 191:114585. [PMID: 36273512 DOI: 10.1016/j.addr.2022.114585] [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: 05/29/2022] [Revised: 09/25/2022] [Accepted: 10/12/2022] [Indexed: 01/24/2023]
Abstract
Despite the advances in immunotherapy for cancer treatment, patients still obtain limited benefits, mostly owing to unrestrained tumour self-expansion and immune evasion that exploits immunoregulatory mechanisms. Traditionally, myeloid cells have a dominantly immunosuppressive role. However, the complicated populations of the myeloid cells and their multilateral interactions with tumour/stromal/lymphoid cells and physical abnormalities in the tumour microenvironment (TME) determine their heterogeneous functions in tumour development and immune response. Tumour-associated myeloid cells (TAMCs) include monocytes, tumour-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), and granulocytes. Single-cell profiling revealed heterogeneous TAMCs composition, sub-types, and transcriptomic signatures across 15 human cancer types. We systematically reviewed the biophysical heterogeneity of TAMC composition and pro/anti-tumoral and immuno-suppressive/stimulating properties of myeloid-derived microenvironments. We also summarised comprehensive clinical strategies to overcome resistance to immunotherapy from three dimensions: targeting TAMCs, reversing physical abnormalities, utilising nanomedicines, and finally, put forward futuristic perspectives for scientific and clinical research.
Collapse
Affiliation(s)
- Jie Zhao
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Yiting Dong
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Yundi Zhang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China.
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
43
|
Hu J, Huang H, Xi Z, Ma S, Ming J, Dong F, Guo H, Zhang H, Zhao E, Yao G, Yang L, Zhang F, Zheng W, Chen H, Huang T, Li L. LncRNA SEMA3B-AS1 inhibits breast cancer progression by targeting miR-3940/KLLN axis. Cell Death Dis 2022; 13:800. [PMID: 36123344 PMCID: PMC9485163 DOI: 10.1038/s41419-022-05189-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 08/07/2022] [Accepted: 08/15/2022] [Indexed: 02/05/2023]
Abstract
Long noncoding RNAs (lncRNAs) play crucial regulatory roles in the progression of various cancers. However, the functional roles of lncRNAs in breast cancer remain unclear. In this study, we investigated the functional role of a novel long noncoding RNA SEMA3B-AS1 (lncRNA SEAS1) in breast cancer progression and the underlying mechanisms. SEAS1 was downregulated in the triple-negative breast cancer (TNBC) tissues compared with the para-carcinoma tissues, which was associated with poor prognosis of TNBC patients. We demonstrated that SEAS1 knockdown significantly increased the proliferation, migration, and invasion of TNBC cell lines, whereas SEAS1 overexpression reversed these effects. Bioinformatics analysis demonstrated that microRNA (miR)-3940-3p was a potential target of SEAS1. Mechanistically, RNA immunoprecipitation (RIP) and luciferase reporter assays confirmed that lncRNA SEMA3B-AS1 acted as sponge for miR-3940-3p, preventing the degradation of its target gene KLLN, which acts as a tumor-inhibiter in TNBC. Moreover, RNA pulldown, mass spectrometry, ChIP, and luciferase reporter assays confirmed that SMAD3 directly interacted with the promoter of SEAS1 and suppressed its transcription, thereby promoting TNBC progression. The clinical samples of TNBC confirmed SEAS1 was correlated inversely with lymphatic and distant metastasis. In conclusion, our findings reveal a novel pathway for TNBC progression via SMAD3/lncRNA SEAS1/miR-3940-3p/KLLN axis, and suggest that SEAS1 may serve as a potential biomarker and therapeutic target for TNBC.
Collapse
Affiliation(s)
- Jin Hu
- grid.33199.310000 0004 0368 7223Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,grid.412632.00000 0004 1758 2270 Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haohao Huang
- grid.417279.eDepartment of Neurosurgery, General Hospital of Central Theater Command of Chinese People’s Liberation Army, Wuhan, 430070 PR China
| | - Zihan Xi
- grid.33199.310000 0004 0368 7223Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Shenghui Ma
- grid.417279.eDepartment of Neurosurgery, General Hospital of Central Theater Command of Chinese People’s Liberation Army, Wuhan, 430070 PR China
| | - Jie Ming
- grid.33199.310000 0004 0368 7223Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Fang Dong
- grid.33199.310000 0004 0368 7223Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Hui Guo
- grid.33199.310000 0004 0368 7223Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Huiqiong Zhang
- grid.33199.310000 0004 0368 7223Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Ende Zhao
- grid.33199.310000 0004 0368 7223Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Guojie Yao
- grid.417279.eDepartment of Neurosurgery, General Hospital of Central Theater Command of Chinese People’s Liberation Army, Wuhan, 430070 PR China
| | - Liu Yang
- grid.417279.eDepartment of Neurosurgery, General Hospital of Central Theater Command of Chinese People’s Liberation Army, Wuhan, 430070 PR China
| | - Feng Zhang
- Department of Emergency Medicine, Affiliated Hospital of Sergeant School Affiliated to Army Medical University, Shijiazhuang, 516562 China
| | - Wuping Zheng
- grid.443397.e0000 0004 0368 7493Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570102 China
| | - Hengyu Chen
- grid.443397.e0000 0004 0368 7493Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570102 China
| | - Tao Huang
- grid.33199.310000 0004 0368 7223Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Lei Li
- grid.33199.310000 0004 0368 7223Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| |
Collapse
|
44
|
Jiang J, Ying H. Revealing the crosstalk between nasopharyngeal carcinoma and immune cells in the tumor microenvironment. J Exp Clin Cancer Res 2022; 41:244. [PMID: 35964134 PMCID: PMC9375932 DOI: 10.1186/s13046-022-02457-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/01/2022] [Indexed: 01/13/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) arises from the epithelial cells located in the nasopharynx and has a distinct geographic distribution. Chronic Epstein-Barr virus (EBV) infection, as its most common causative agents, can be detected in 100% of NPC types. In-depth studies of the cellular and molecular events leading to immunosuppression in NPC have revealed new therapeutic targets and diverse combinations that promise to benefit patients with highly refractory, advanced and metastatic NPC. This paper reviews the mechanisms by which NPC cells to circumvent immune surveillance and approaches being attempted to restore immunity. We integrate existing insights into anti-NPC immunity and molecular signaling pathways as well as targeting therapies in anticipation of broader applicability and effectiveness in advanced metastatic NPC.
Collapse
|
45
|
Glathar AR, Oyelakin A, Gluck C, Bard J, Sinha S. p63 Directs Subtype-Specific Gene Expression in HPV+ Head and Neck Squamous Cell Carcinoma. Front Oncol 2022; 12:879054. [PMID: 35712470 PMCID: PMC9192977 DOI: 10.3389/fonc.2022.879054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/19/2022] [Indexed: 11/22/2022] Open
Abstract
The complex heterogeneity of head and neck squamous cell carcinoma (HNSCC) reflects a diverse underlying etiology. This heterogeneity is also apparent within Human Papillomavirus-positive (HPV+) HNSCC subtypes, which have distinct gene expression profiles and patient outcomes. One aggressive HPV+ HNSCC subtype is characterized by elevated expression of genes involved in keratinization, a process regulated by the oncogenic transcription factor ΔNp63. Furthermore, the human TP63 gene locus is a frequent HPV integration site and HPV oncoproteins drive ΔNp63 expression, suggesting an unexplored functional link between ΔNp63 and HPV+ HNSCC. Here we show that HPV+ HNSCCs can be molecularly stratified according to ΔNp63 expression levels and derive a ΔNp63-associated gene signature profile for such tumors. We leveraged RNA-seq data from p63 knockdown cells and ChIP-seq data for p63 and histone marks from two ΔNp63high HPV+ HNSCC cell lines to identify an epigenetically refined ΔNp63 cistrome. Our integrated analyses reveal crucial ΔNp63-bound super-enhancers likely to mediate HPV+ HNSCC subtype-specific gene expression that is anchored, in part, by the PI3K-mTOR pathway. These findings implicate ΔNp63 as a key regulator of essential oncogenic pathways in a subtype of HPV+ HNSCC that can be exploited as a biomarker for patient stratification and treatment choices.
Collapse
|
46
|
Xie X, Lee J, Iwase T, Kai M, Ueno NT. Emerging drug targets for triple-negative breast cancer: A guided tour of the preclinical landscape. Expert Opin Ther Targets 2022; 26:405-425. [PMID: 35574694 DOI: 10.1080/14728222.2022.2077188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is the most fatal molecular subtype of breast cancer because of its aggressiveness and resistance to chemotherapy. FDA-approved therapies for TNBC are limited to poly(ADP-ribose) polymerase inhibitors, immune checkpoint inhibitors, and trophoblast cell surface antigen 2-targeted antibody-drug conjugate. Therefore, developing a novel effective targeted therapy for TNBC is an urgent unmet need. AREAS COVERED In this narrative review, we discuss emerging targets for TNBC treatment discovered in early translational studies. We focus on cancer cell membrane molecules, hyperactive intracellular signaling pathways, and the tumor microenvironment (TME) based on their druggability, therapeutic potency, specificity to TNBC, and application in immunotherapy. EXPERT OPINION The significant challenges in the identification and validation of TNBC-associated targets are 1) application of appropriate genetic, molecular, and immunological approaches for modulating the target, 2) establishment of a proper mouse model that accurately represents the human immune TME, 3) TNBC molecular heterogeneity, and 4) failure translation of preclinical findings to clinical practice. To overcome those difficulties, future research needs to apply novel technology, such as single-cell RNA sequencing, thermostable group II intron reverse transcriptase sequencing, and humanized mouse models. Further, combination treatment targeting multiple pathways in both the TNBC tumor and its TME is essential for effective disease control.
Collapse
Affiliation(s)
- Xuemei Xie
- Section of Translational Breast Cancer Research, Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jangsoon Lee
- Section of Translational Breast Cancer Research, Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Toshiaki Iwase
- Section of Translational Breast Cancer Research, Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Megumi Kai
- Section of Translational Breast Cancer Research, Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research, Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
47
|
Blaye C, Boyer T, Peyraud F, Domblides C, Larmonier N. Beyond Immunosuppression: The Multifaceted Functions of Tumor-Promoting Myeloid Cells in Breast Cancers. Front Immunol 2022; 13:838040. [PMID: 35309358 PMCID: PMC8927658 DOI: 10.3389/fimmu.2022.838040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/02/2022] [Indexed: 11/20/2022] Open
Abstract
Breast cancers are commonly associated with an immunosuppressive microenvironment responsible for tumor escape from anti-cancer immunity. Cells of the myeloid lineage account for a major part of this tumor-promoting landscape. These myeloid cells are composed of heterogeneous subsets at different stages of differentiation and have traditionally been described by their cardinal ability to suppress innate and adaptive anticancer immunity. However, evidence has accumulated that, beyond their immunosuppressive properties, breast cancer-induced myeloid cells are also equipped with a broad array of “non-immunological” tumor-promoting functions. They therefore represent major impediments for anticancer therapies, particularly for immune-based interventions. We herein analyze and discuss current literature related to the versatile properties of the different myeloid cell subsets engaged in breast cancer development. We critically assess persisting difficulties and challenges in unequivocally discriminate dedicated subsets, which has so far prevented both the selective targeting of these immunosuppressive cells and their use as potential biomarkers. In this context, we propose the concept of IMCGL, “pro-tumoral immunosuppressive myeloid cells of the granulocytic lineage”, to more accurately reflect the contentious nature and origin of granulocytic cells in the breast tumor microenvironment. Future research prospects related to the role of this myeloid landscape in breast cancer are further considered.
Collapse
Affiliation(s)
- Céline Blaye
- Centre National de la Recherche Scientific (CNRS) Unité Mixte de Recherche (UMR) 5164, ImmunoConcEpT, Bordeaux, France.,Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Thomas Boyer
- Centre National de la Recherche Scientific (CNRS) Unité Mixte de Recherche (UMR) 5164, ImmunoConcEpT, Bordeaux, France
| | - Florent Peyraud
- Centre National de la Recherche Scientific (CNRS) Unité Mixte de Recherche (UMR) 5164, ImmunoConcEpT, Bordeaux, France
| | - Charlotte Domblides
- Centre National de la Recherche Scientific (CNRS) Unité Mixte de Recherche (UMR) 5164, ImmunoConcEpT, Bordeaux, France.,Service d'Oncologie Médicale, Centre Hospitalo-Universitaire (CHU) Bordeaux, Bordeaux, France
| | - Nicolas Larmonier
- Centre National de la Recherche Scientific (CNRS) Unité Mixte de Recherche (UMR) 5164, ImmunoConcEpT, Bordeaux, France.,Department of Biological and Medical Sciences, University of Bordeaux, Bordeaux, France
| |
Collapse
|
48
|
Yu S, Li D, Zhang N, Ni S, Sun M, Wang L, Xiao H, Liu D, Liu J, Yu Y, Zhang Z, Yeung STY, Zhang S, Lu A, Zhang Z, Zhang B, Zhang G. Drug discovery of sclerostin inhibitors. Acta Pharm Sin B 2022; 12:2150-2170. [PMID: 35646527 PMCID: PMC9136615 DOI: 10.1016/j.apsb.2022.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/03/2021] [Accepted: 12/16/2021] [Indexed: 12/18/2022] Open
Abstract
Sclerostin, a protein secreted from osteocytes, negatively regulates the WNT signaling pathway by binding to the LRP5/6 co-receptors and further inhibits bone formation and promotes bone resorption. Sclerostin contributes to musculoskeletal system-related diseases, making it a promising therapeutic target for the treatment of WNT-related bone diseases. Additionally, emerging evidence indicates that sclerostin contributes to the development of cancers, obesity, and diabetes, suggesting that it may be a promising therapeutic target for these diseases. Notably, cardiovascular diseases are related to the protective role of sclerostin. In this review, we summarize three distinct types of inhibitors targeting sclerostin, monoclonal antibodies, aptamers, and small-molecule inhibitors, from which monoclonal antibodies have been developed. As the first-in-class sclerostin inhibitor approved by the U.S. FDA, the monoclonal antibody romosozumab has demonstrated excellent effectiveness in the treatment of postmenopausal osteoporosis; however, it conferred high cardiovascular risk in clinical trials. Furthermore, romosozumab could only be administered by injection, which may cause compliance issues for patients who prefer oral therapy. Considering these above safety and compliance concerns, we therefore present relevant discussion and offer perspectives on the development of next-generation sclerostin inhibitors by following several ways, such as concomitant medication, artificial intelligence-based strategy, druggable modification, and bispecific inhibitors strategy.
Collapse
|
49
|
Chung DC, Jacquelot N, Ghaedi M, Warner K, Ohashi PS. Innate Lymphoid Cells: Role in Immune Regulation and Cancer. Cancers (Basel) 2022; 14:2071. [PMID: 35565201 PMCID: PMC9102917 DOI: 10.3390/cancers14092071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
Immune regulation is composed of a complex network of cellular and molecular pathways that regulate the immune system and prevent tissue damage. It is increasingly clear that innate lymphoid cells (ILCs) are also armed with immunosuppressive capacities similar to well-known immune regulatory cells (i.e., regulatory T cells). In cancer, immunoregulatory ILCs have been shown to inhibit anti-tumour immune response through various mechanisms including: (a) direct suppression of anti-tumour T cells or NK cells, (b) inhibiting T-cell priming, and (c) promoting other immunoregulatory cells. To provide a framework of understanding the role of immunosuppressive ILCs in the context of cancer, we first outline a brief history and challenges related to defining immunosuppressive ILCs. Furthermore, we focus on the mechanisms of ILCs in suppressing anti-tumour immunity and consequentially promoting tumour progression.
Collapse
Affiliation(s)
- Douglas C. Chung
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Nicolas Jacquelot
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Maryam Ghaedi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Kathrin Warner
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Pamela S. Ohashi
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| |
Collapse
|
50
|
Wang Y, Lu H, Wang Z, Li Y, Chen X. TGF-β1 Promotes Autophagy and Inhibits Apoptosis in Breast Cancer by Targeting TP63. Front Oncol 2022; 12:865067. [PMID: 35480110 PMCID: PMC9035888 DOI: 10.3389/fonc.2022.865067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background Breast cancer (BC) is a prevalent female cancer, which has high morbidity and mortality. However, the pathogenesis of BC has not been fully elucidated. Studies have shown that TGF-β1 plays an important role in regulating the balance between autophagy and apoptosis of tumor. We aim to clarify the specific mechanism of autophagy and apoptosis in breast cancer maintaining the tumor microenvironment. Methods The clinical characteristics of 850 BC patients were retrieved from the TCGA database. Differentially expressed autophagy-related genes (DEARGs) between tumor and normal tissues were obtained by the Wilcox test. Through Cox proportional hazard regression analysis, the prognostic risk model was constructed and verified by the ROC curve. We used MDC staining, colony formation assay, CCK-8, flow cytometric analysis to confirm the importance of TGF-β1 on the autophagy and apoptosis of breast cancer cells. Furthermore, western blot was performed to determine the relative expression of protein. The Kaplan-Meier Plotter database was utilized to identify the prognostic value of TP63. Results We successfully constructed a prognostic risk model of breast cancer and screened out an autophagy-related prognostic gene -TP63. We predicted that TGF-β1 and TP63 have a binding site in the JASPAR database as expected. Additionally, TGF-β1 promoted autophagy and inhibited apoptosis of breast cancer cells by inhibiting the expression of TP63. Conclusion Our study demonstrated that the molecular mechanism of TGF-β/TP63 signaling in regulating autophagy and apoptosis of breast cancer and provided a potential prognostic marker in breast cancer.
Collapse
Affiliation(s)
- Yichao Wang
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou Univesity Hospital), Taizhou, China
| | - Hongsheng Lu
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou Univesity Hospital), Taizhou, China
| | - Zhongrong Wang
- Key Laboratory of Brain-Like Neuromorphic Devices and Systems of Hebei Province College of Electron and Information Engineering, Hebei University, Baoding, China
| | - Yueguo Li
- Department of Clinical Laboratory, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiaoying Chen
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou Univesity Hospital), Taizhou, China
| |
Collapse
|