1
|
Zhou P, Li Z, Liu F, Kwon E, Hsieh TC, Ye S, Vasudevan S, Lee JA, Tran KV, Zhou C. BAMBI integrates biostatistical and artificial intelligence methods to improve RNA biomarker discovery. Brief Bioinform 2025; 26:bbaf073. [PMID: 40121554 PMCID: PMC11929966 DOI: 10.1093/bib/bbaf073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/09/2025] [Accepted: 01/26/2025] [Indexed: 03/25/2025] Open
Abstract
RNA biomarkers enable early and precise disease diagnosis, monitoring, and prognosis, facilitating personalized medicine and targeted therapeutic strategies. However, identification of RNA biomarkers is hindered by the challenge of analyzing relatively small yet high-dimensional transcriptomics datasets, typically comprising fewer than 1000 biospecimens but encompassing hundreds of thousands of RNAs, especially noncoding RNAs. This complexity leads to several limitations in existing methods, such as poor reproducibility on independent datasets, inability to directly process omics data, and difficulty in identifying noncoding RNAs as biomarkers. Additionally, these methods often yield results that lack biological interpretation and clinical utility. To overcome these challenges, we present BAMBI (Biostatistical and Artificial-intelligence Methods for Biomarker Identification), a computational tool integrating biostatistical approaches and machine-learning algorithms. By initially reducing high dimensionality through biologically informed statistical methods followed by machine learning-based feature selection, BAMBI significantly enhances the accuracy and clinical utility of identified RNA biomarkers and also includes noncoding RNA biomarkers that existing methods may overlook. BAMBI outperformed existing methods on both real and simulated datasets by identifying individual and panel biomarkers with fewer RNAs while still ensuring superior prediction accuracy. BAMBI was benchmarked on multiple transcriptomics datasets across diseases, including breast cancer, psoriasis, and leukemia. The prognostic biomarkers for acute myeloid leukemia discovered by BAMBI showed significant correlations with patient survival rates in an independent cohort, highlighting its potential for enhancing clinical outcomes. The software is available on GitHub (https://github.com/CZhouLab/BAMBI).
Collapse
Affiliation(s)
- Peng Zhou
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Zixiu Li
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Feifan Liu
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Euijin Kwon
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Tien-Chan Hsieh
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Shangyuan Ye
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health and Science University, 2720 S Moody Ave, Portland, OR 97201, United States
| | - Shobha Vasudevan
- Brown RNA Center, Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02903, United States
| | - Jung Ae Lee
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Khanh-Van Tran
- Division of Cardiology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Chan Zhou
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
- The RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
- UMass Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| |
Collapse
|
2
|
Moura T, Laranjeira P, Caramelo O, Gil AM, Paiva A. Breast Cancer and Tumor Microenvironment: The Crucial Role of Immune Cells. Curr Oncol 2025; 32:143. [PMID: 40136347 PMCID: PMC11941043 DOI: 10.3390/curroncol32030143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
Breast cancer is the most common type of cancer in women and the second leading cause of death by cancer. Despite recent advances, the mortality rate remains high, underlining the need to develop new therapeutic approaches. The complex interaction between cancer cells and the tumor microenvironment (TME) is crucial in determining tumor progression, therapy response, and patient prognosis. Understanding the role of immune cells in carcinogenesis and tumor progression can help improve targeted therapeutic options, increasing the likelihood of a favorable prognosis. Therefore, this review aims to critically analyze the complex interaction between tumor cells and immune cells, emphasizing the clinical and therapeutic implications. Additionally, we explore advances in immunotherapies, with a focus on immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Tânia Moura
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Paula Laranjeira
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Group of Environmental Genetics of Oncobiology (CIMAGO), Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Center of Neurosciences and Cell (CNC), University of Coimbra, 3000-504 Coimbra, Portugal
| | - Olga Caramelo
- Gynecology Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-075 Coimbra, Portugal;
| | - Ana M. Gil
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Group of Environmental Genetics of Oncobiology (CIMAGO), Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Ciências Biomédicas Laboratoriais, Instituto Politécnico de Coimbra, ESTESC—Coimbra Health School, 3046-854 Coimbra, Portugal
| |
Collapse
|
3
|
Wang Z, Dai Y, Zhou Y, Wang Y, Chen P, Li Y, Zhang Y, Wang X, Hu Y, Li H, Li G, Jing Y. Research progress of T cells in cholangiocarcinoma. Front Immunol 2025; 16:1453344. [PMID: 40070825 PMCID: PMC11893616 DOI: 10.3389/fimmu.2025.1453344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Cholangiocarcinoma (CCA), a malignant tumor, is typically challenging to detect early and often results in a poor prognosis. In recent years, research interest has grown in the potential application of immunotherapy for CCA treatment. T cells, as a crucial component of the immune system, play a significant role in immune surveillance and therapy for cholangiocarcinoma. This article provides a review of the research advancements concerning T cells in cholangiocarcinoma patients, including their distribution, functional status, and correlation with patient prognosis within the tumor microenvironment. It further discusses the potential applications and challenges of immunotherapy strategies targeting T cells in CCA treatment and anticipates future research directions. A more profound understanding of T cells' role in cholangiocarcinoma can guide the development of clinical treatment strategies, thereby enhancing patient survival rates and quality of life. Finally, we explored the potential risks and side effects of immunotherapy for T-cell cholangiocarcinoma.
Collapse
Affiliation(s)
- Zhiming Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yunyan Dai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yunpeng Zhou
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yi Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Pinggui Chen
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yaoxuan Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yunfei Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xiaocui Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Ying Hu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Haonan Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Gaopeng Li
- Department of Hepatobiliary Surgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yukai Jing
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
4
|
Wang B, Xia Y, Zhou C, Zeng Y, Son HG, Demehri S. CD4+ T helper 2 cell-macrophage crosstalk induces IL-24-mediated breast cancer suppression. JCI Insight 2025; 10:e180962. [PMID: 39782693 PMCID: PMC11721301 DOI: 10.1172/jci.insight.180962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 11/08/2024] [Indexed: 01/12/2025] Open
Abstract
CD4+ T cells contribute to antitumor immunity and are implicated in the efficacy of cancer immunotherapies. In particular, CD4+ T helper 2 (Th2) cells were recently found to block spontaneous breast carcinogenesis. However, the antitumor potential of Th2 cells in targeting established breast cancer remains uncertain. Herein, we demonstrate that Th2 cells induced by the topical calcipotriol/thymic stromal lymphopoietin cytokine axis suppressed the growth of established mammary tumors in mice. Interleukin-24 (IL-24), an anticancer cytokine, was highly upregulated in macrophages infiltrating calcipotriol-treated mammary tumors. Macrophages expressed IL-24 in response to IL-4 signaling in combination with Toll-like receptor 4 (TLR4) agonists (e.g., HMGB1) in vitro. Calcipotriol treatment significantly increased HMGB1 release by tumor cells in vivo. CD4+ T cell depletion reduced HMGB1 and IL-24 expression, reversing calcipotriol's therapeutic efficacy. Macrophage depletion and TLR4 inhibition also reduced the therapeutic efficacy of calcipotriol. Importantly, calcipotriol treatment failed to control mammary tumors lacking the IL-24 receptor on tumor cells. Collectively, our findings reveal that Th2 cell-macrophage crosstalk leads to IL-24-mediated tumor cell death, highlighting a promising therapeutic strategy to tackle breast cancer.
Collapse
Affiliation(s)
- Bo Wang
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Urology and
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yun Xia
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Can Zhou
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yuhan Zeng
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Heehwa G. Son
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Oka T, Smith SS, Son HG, Lee T, Oliver-Garcia VS, Mortaja M, Trerice KE, Isakoff LS, Conrad DN, Azin M, Raval NS, Tabacchi M, Emdad L, Das SK, Fisher PB, Cornelius LA, Demehri S. T helper 2 cell-directed immunotherapy eliminates precancerous skin lesions. J Clin Invest 2025; 135:e183274. [PMID: 39744942 PMCID: PMC11684800 DOI: 10.1172/jci183274] [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: 05/28/2024] [Accepted: 10/08/2024] [Indexed: 01/06/2025] Open
Abstract
The continuous rise in skin cancer incidence highlights an imperative for improved skin cancer prevention. Topical calcipotriol-plus-5-fluorouracil (calcipotriol-plus-5-FU) immunotherapy effectively eliminates precancerous skin lesions and prevents squamous cell carcinoma (SCC) in patients. However, its mechanism of action remains unclear. Herein, we demonstrate that calcipotriol-plus-5-FU immunotherapy induces T helper type 2 (Th2) immunity, eliminating premalignant keratinocytes in humans. CD4+ Th2 cells were required and were sufficient downstream of thymic stromal lymphopoietin cytokine induction by calcipotriol to suppress skin cancer development. Th2-associated cytokines induced IL-24 expression in cancer cells, resulting in toxic autophagy and anoikis followed by apoptosis. Calcipotriol-plus-5-FU immunotherapy was dependent on IL-24 to suppress skin carcinogenesis in vivo. Collectively, our findings establish a critical role for Th2 immunity in cancer immunoprevention and highlight the Th2/IL-24 axis as an innovative target for skin cancer prevention and therapy.
Collapse
Affiliation(s)
- Tomonori Oka
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sabrina S. Smith
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Heehwa G. Son
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Truelian Lee
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Valeria S. Oliver-Garcia
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mahsa Mortaja
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kathryn E. Trerice
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lily S. Isakoff
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Danielle N. Conrad
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marjan Azin
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Neel S. Raval
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mary Tabacchi
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Swadesh K. Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Paul B. Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Lynn A. Cornelius
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Krantz Family Center for Cancer Research and Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Hargrove-Wiley E, Obodo D, Bindeman W, Fingleton B. Elucidating Sex-Specific Immune Profiles in a Breast Cancer Model. Int J Mol Sci 2024; 25:13113. [PMID: 39684829 DOI: 10.3390/ijms252313113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/23/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Breast cancer is commonly thought of as a "women's disease". However, men are increasingly diagnosed with the disease, and their mortality rates are disparately higher than those of female patients. The abundance and composition of the immune microenvironment are determinants of breast cancer progression and survival. It is well documented that there are sex-specific differences in the immune response to several diseases, including various cancers. However, the effects of these differences in the context of breast cancer remain to be explored. This study demonstrates sex differences in the hormonal and immune landscape of the MMTV-PyMT transgenic murine model of female and male ER+ breast cancer using single-cell RNA sequencing (scRNA-Seq), whole-slide immunohistochemistry, and flow cytometry. Mammary tumors of transgenic male mice had increased estrogen receptor alpha expression and enriched nuclear binding signatures compared to female tumors. In the tumor immune compartment, male mice had lower intratumoral leukocyte infiltration. Yet, scRNA-Seq analysis reveals a more immunostimulatory microenvironment and increased antitumor immune populations in the primary and metastatic lungs as compared to transgenic females. Despite a more favorable innate immune profile, the metastatic burden was increased in male mice. Our data support a sex-dependent immune response in mammary carcinoma associated with the tumor, and likely host, hormonal environment. With emerging therapeutics targeting the tumor immune microenvironment, characterizing immune profiles is critical for optimizing their use in all breast cancer patients.
Collapse
Affiliation(s)
- Ebony Hargrove-Wiley
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Dora Obodo
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Wendy Bindeman
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Barbara Fingleton
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
7
|
Zhang Y, Qin X, Lou W, Wang L, Lu W, Gao C, Hu S. Deciphering the cellular landscape and potential targets of nasopharyngeal and oral cancers using single-cell RNA sequencing. Cell Biol Int 2024; 48:1849-1861. [PMID: 39205595 DOI: 10.1002/cbin.12236] [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: 02/08/2024] [Revised: 07/16/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Cellular heterogeneity in nasopharyngeal cancer (NPC) and oral cancer remains unclear. In the current study, using single-cell RNA sequencing techniques, we investigated the cellular landscape in NPC and oral cancers. We identified a diverse range of cell types within the tumor microenvironment (TME) and variations in cell infiltration between NPC and oral cancer. In oral cancer, we observed a predominant infiltration of epithelial cells, fibroblasts, and endothelial cells (ECs), while T cells were the main infiltrating cell population in NPCs. We further classified these infiltrating cells into subclusters. Additionally, we observed complex interactions among cells that led to distinct trajectories. In particular, a unique epithelial subcluster with high expression of major histocompatibility complex class II (MHC-II) molecules was correlated with a favorable outcome and infiltration of CD4+ T cells. In addition, MHC-II+ epithelial cells inhibited mouse tumor growth and promoted T-cell infiltration. Consequently, our findings provide a deep understanding of the TME showing a significant prognostic value and therapeutic potential.
Collapse
Affiliation(s)
- Yanfei Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoyu Qin
- Department of Vascular Surgery, Zhengzhou Central Hospital, Zhengzhou, Henan, China
| | - Weihua Lou
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liang Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wuhao Lu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Changhui Gao
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shousen Hu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Bhamidipati P, Nagaraju GP, Malla R. Immunoglobulin-binding protein and Toll-like receptors in immune landscape of breast cancer. Life Sci 2024; 358:123196. [PMID: 39481836 DOI: 10.1016/j.lfs.2024.123196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/29/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Breast cancer (BC) is a complex disease exhibiting significant heterogeneity and encompassing various molecular subtypes. Among these, triple-negative breast cancer (TNBC) stands out as one of the most challenging types, characterized by its aggressive nature and poor prognosis. This review embarks on a comprehensive exploration of the immune landscape of BC, with a primary focus on the functional and structural characterization of immunoglobulin-binding protein (BiP) and its pivotal role in regulating the unfolded response (UPR) pathway of proteins. Moreover, we unravel the multifaceted functions of BiP in BC, with a special emphasis on the involvement of cell surface BiP in TNBC metastasis, drug resistance, and its contribution to the formation of the tumor microenvironment (TME). We also provide mechanistic insights into how ER-resident BiP mediates the sensitization of drug-resistant BC to different treatment strategies, thereby offering promising avenues for therapeutic intervention. We also delve into the role of Toll-like receptors (TLRs), shedding light on their diverse expression patterns across BC and their influence on modulating the tumor immune response. Understanding the interplay between BiP, TLRs, and the immune response, especially in TNBC, opens avenues for novel immunotherapies. Future research should focus on developing targeted therapies that activate ER-resident BiP or inhibit cell surface BiP, and modulate TLR signaling. Moreover, exploring BiP as a biomarker for TNBC diagnosis, prognosis, and treatment response will be crucial for personalized medicine.
Collapse
Affiliation(s)
- Priyamvada Bhamidipati
- Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - RamaRao Malla
- Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India.
| |
Collapse
|
9
|
Sheng Z, Wang X, Zheng Y, Duan W, Cui J, Gu L, Gao X, Ma J, Cui M, Luo H, Wang W, Shi L, Li H, Zhang B. Genome-wide characterization of extrachromosomal circular DNA in breast cancer and its potential role in carcinogenesis and cancer progression. Cell Rep 2024; 43:114845. [PMID: 39418165 DOI: 10.1016/j.celrep.2024.114845] [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: 02/27/2024] [Revised: 07/01/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Extrachromosomal circular DNAs (eccDNAs) are defined as distinct genomic entities of circular and mobile DNA molecules, but their molecular functions in and impact on breast cancer (BC) are rarely known. This study used Circle-seq to analyze eccDNAs from 19 BC tissues and 17 adjacent normal tissues. We found that eccDNAs are present on all chromosomes and enriched in seven eccDNA hotspot genes (HSGs) associated with the BC pathway. Several eccDNAs harboring entire genes (eccGenes) and eccDNAs harboring miRNAs (eccMIRs) were identified and linked to cancer-relevant pathways. Synthetic eccMIR6748, eccMIR6508, and eccMIR3142 elevated miRNA expression in MCF-7 cells, with eccMIR6748 promoting BC cell migration and invasion by upregulating miR-6748, which suppresses tumor suppressor candidate factor 5 (TUSC5) at the post-transcriptional level. eccMIR6748 also influences BC progression via the p38 mitogen-activated protein kinase (MAPK) signaling pathway. These findings suggest that eccDNAs, which contain functional genomic segments, play a role in BC initiation and progression, offering a dynamic source of genomic plasticity and potential as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Zhimei Sheng
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang, China; Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Xuejie Wang
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Yuanhang Zheng
- Department of Pathology, Weifang People's Hospital, Weifang, Shandong, China
| | - Wanli Duan
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Jiayu Cui
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Lihui Gu
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Xinxin Gao
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Jing Ma
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Meimei Cui
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Hao Luo
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Wenhao Wang
- Department of Medical Oncology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Lihong Shi
- Department of Pharmacology, Shandong Second Medical University, Weifang, Shandong, China
| | - Hongli Li
- Department of Medicine Research Center, Shandong Second Medical University, Weifang, Shandong, China
| | - Baogang Zhang
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang, China.
| |
Collapse
|
10
|
Arpinati L, Carradori G, Scherz-Shouval R. CAF-induced physical constraints controlling T cell state and localization in solid tumours. Nat Rev Cancer 2024; 24:676-693. [PMID: 39251836 DOI: 10.1038/s41568-024-00740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/11/2024]
Abstract
Solid tumours comprise cancer cells that engage in continuous interactions with non-malignant cells and with acellular components, forming the tumour microenvironment (TME). The TME has crucial and diverse roles in tumour progression and metastasis, and substantial efforts have been dedicated into understanding the functions of different cell types within the TME. These efforts highlighted the importance of non-cell-autonomous signalling in cancer, mediating interactions between the cancer cells, the immune microenvironment and the non-immune stroma. Much of this non-cell-autonomous signalling is mediated through acellular components of the TME, known as the extracellular matrix (ECM), and controlled by the cells that secrete and remodel the ECM - the cancer-associated fibroblasts (CAFs). In this Review, we delve into the complex crosstalk among cancer cells, CAFs and immune cells, highlighting the effects of CAF-induced ECM remodelling on T cell functions and offering insights into the potential of targeting ECM components to improve cancer therapies.
Collapse
Affiliation(s)
- Ludovica Arpinati
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Giulia Carradori
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
11
|
Goodin DA, Chau E, Zheng J, O’Connell C, Tiwari A, Xu Y, Niravath P, Chen SH, Godin B, Frieboes HB. Characterization of the Breast Cancer Liver Metastasis Microenvironment via Machine Learning Analysis of the Primary Tumor Microenvironment. CANCER RESEARCH COMMUNICATIONS 2024; 4:2846-2857. [PMID: 39373616 PMCID: PMC11525956 DOI: 10.1158/2767-9764.crc-24-0263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/16/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Breast cancer liver metastases (BCLM) are hypovascular lesions that resist intravenously administered therapies and have grim prognosis. Immunotherapeutic strategies targeting BCLM critically depend on the tumor microenvironment (TME), including tumor-associated macrophages. However, a priori characterization of the BCLM TME to optimize therapy is challenging because BCLM tissue is rarely collected. In contrast to primary breast tumors for which tissue is usually obtained and histologic analysis performed, biopsies or resections of BCLM are generally discouraged due to potential complications. This study tested the novel hypothesis that BCLM TME characteristics could be inferred from the primary tumor tissue. Matched primary and metastatic human breast cancer samples were analyzed by imaging mass cytometry, identifying 20 shared marker clusters denoting macrophages (CD68, CD163, and CD206), monocytes (CD14), immune response (CD56, CD4, and CD8a), programmed cell death protein 1, PD-L1, tumor tissue (Ki-67 and phosphorylated ERK), cell adhesion (E-cadherin), hypoxia (hypoxia-inducible factor-1α), vascularity (CD31), and extracellular matrix (alpha smooth muscle actin, collagen, and matrix metalloproteinase 9). A machine learning workflow was implemented and trained on primary tumor clusters to classify each metastatic cluster density as being either above or below median values. The proposed approach achieved robust classification of BCLM marker data from matched primary tumor samples (AUROC ≥ 0.75, 95% confidence interval ≥ 0.7, on the validation subsets). Top clusters for prediction included CD68+, E-cad+, CD8a+PD1+, CD206+, and CD163+MMP9+. We conclude that the proposed workflow using primary breast tumor marker data offers the potential to predict BCLM TME characteristics, with the longer term goal to inform personalized immunotherapeutic strategies targeting BCLM. SIGNIFICANCE BCLM tissue characterization to optimize immunotherapy is difficult because biopsies or resections are rarely performed. This study shows that a machine learning approach offers the potential to infer BCLM characteristics from the primary tumor tissue.
Collapse
Affiliation(s)
- Dylan A. Goodin
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Eric Chau
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Junjun Zheng
- Immunomonitoring Core, Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, Texas
| | - Cailin O’Connell
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Anjana Tiwari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
| | - Yitian Xu
- Immunomonitoring Core, Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, Texas
| | - Polly Niravath
- Breast Medical Oncology Faculty, Houston Methodist Cancer Center, Houston, Texas
| | - Shu-Hsia Chen
- Immunomonitoring Core, Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, Texas
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, New York
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - Hermann B. Frieboes
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
- UofL Health – Brown Cancer Center, University of Louisville, Louisville, Kentucky
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
12
|
Xu H, Du W, Jing X, Xie J, Li P. Development of a prognostic model for lung adenocarcinoma polarity-related genes and analysis of immune landscape. Biotechnol Appl Biochem 2024; 71:817-834. [PMID: 38475658 DOI: 10.1002/bab.2579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Despite the progress made in the management of lung adenocarcinoma (LUAD), the overall prognosis for LUAD individuals remains suboptimal. While the role of cell polarity in tumor invasion and metastasis is well established, its prognostic significance in LUAD is still unknown. Differential analysis was performed on the Cancer Genome Atlas (TCGA)-LUAD and normal lung tissue, and candidate genes were identified by intersecting differentially expressed genes with polarity-related genes (PRGs). A prognostic model was constructed using univariate and multivariate Cox regression and LASSO regression. To enhance the robustness of the analysis, an independent prognostic analysis was conducted by incorporating relevant clinical information. The accuracy and sensitivity of the model were validated using survival analysis and ROC curves. Finally, immune landscape, immune therapy, tumor mutation burden, and drug sensitivity analysis were carried out on high- and low-risk patients. Ten prognostic genes were screened to divide LUAD patients into different risk groups. Survival analysis, ROC curves, and univariate/multivariate Cox regression analyses collectively demonstrated the favorable predictive performance of the model, which could be an independent prognostic factor. The nomogram, in conjunction with the calibration curve, demonstrated the model's compelling predictive capacity in prognosticating the overall survival of LUAD individuals. Low-risk LUAD patients exhibited heightened levels of immune cell infiltration, immune scores, and immune checkpoint expression compared to high-risk individuals. So, they may have a greater likelihood of benefiting from immune therapy. The high-risk group demonstrated a remarkably higher tumor mutation burden (TMB) in contrast with the low-risk group. XAV-939, Fulvestrant, and SR16157 may have potential value in the clinical use of LUAD. We revealed the potential linkage between PRGs and LUAD prognosis, and the application of these prognostic factors in risk stratification and prognosis prediction of LUAD patients may be of great significance.
Collapse
Affiliation(s)
- Hongqiu Xu
- Department of General Medicine, Huai'an Hospital of Huai'an City, Huai'an City, Jiangsu Province, China
| | - Wenqiang Du
- Department of General Medicine, Huai'an Hospital of Huai'an City, Huai'an City, Jiangsu Province, China
| | - Xuelong Jing
- Department of General Medicine, Huai'an Hospital of Huai'an City, Huai'an City, Jiangsu Province, China
| | - Jingen Xie
- Department of General Medicine, Huai'an Hospital of Huai'an City, Huai'an City, Jiangsu Province, China
| | - Pengfei Li
- Department of General Medicine, Huai'an Hospital of Huai'an City, Huai'an City, Jiangsu Province, China
| |
Collapse
|
13
|
Wang Z, Tang R, Wang H, Li X, Liu Z, Li W, Peng G, Zhou H. Bioinformatics analysis of the role of lysosome-related genes in breast cancer. Comput Methods Biomech Biomed Engin 2024:1-20. [PMID: 39054687 DOI: 10.1080/10255842.2024.2379936] [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: 03/12/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024]
Abstract
This study aimed to investigate the roles of lysosome-related genes in BC prognosis and immunity. Transcriptome data from TCGA and MSigDB, along with lysosome-related gene sets, underwent NMF cluster analysis, resulting in two subtypes. Using lasso regression and univariate/multivariate Cox regression analysis, an 11-gene signature was successfully identified and verified. High- and low-risk populations were dominated by HR+ sample types. There were differences in pathway enrichment, immune cell infiltration, and immune scores. Sensitive drugs targeting model genes were screened using GDSC and CCLE. This study constructed a reliable prognostic model with lysosome-related genes, providing valuable insights for BC clinical immunotherapy.
Collapse
Affiliation(s)
- Zhongming Wang
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Ruiyao Tang
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Huazhong Wang
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Xizhang Li
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Zhenbang Liu
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Wenjie Li
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Gui Peng
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Huaiying Zhou
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| |
Collapse
|
14
|
Gerashchenko T, Frolova A, Patysheva M, Fedorov A, Stakheyeva M, Denisov E, Cherdyntseva N. Breast Cancer Immune Landscape: Interplay Between Systemic and Local Immunity. Adv Biol (Weinh) 2024; 8:e2400140. [PMID: 38727796 DOI: 10.1002/adbi.202400140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/16/2024] [Indexed: 07/13/2024]
Abstract
Breast cancer (BC) is one of the most common malignancies in women worldwide. Numerous studies in immuno-oncology and successful trials of immunotherapy have demonstrated the causal role of the immune system in cancer pathogenesis. The interaction between the tumor and the immune system is known to have a dual nature. Despite cytotoxic lymphocyte activity against transformed cells, a tumor can escape immune surveillance and leverage chronic inflammation to maintain its own development. Research on antitumor immunity primarily focuses on the role of the tumor microenvironment, whereas the systemic immune response beyond the tumor site is described less thoroughly. Here, a comprehensive review of the formation of the immune profile in breast cancer patients is offered. The interplay between systemic and local immune reactions as self-sustaining mechanism of tumor progression is described and the functional activity of the main cell populations related to innate and adaptive immunity is discussed. Additionally, the interaction between different functional levels of the immune system and their contribution to the development of the pro- or anti-tumor immune response in BC is highlighted. The presented data can potentially inform the development of new immunotherapy strategies in the treatment of patients with BC.
Collapse
Affiliation(s)
- Tatiana Gerashchenko
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Anastasia Frolova
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
- Tomsk State University, 36 Lenin Ave., Tomsk, 634050, Russia
| | - Marina Patysheva
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Anton Fedorov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Marina Stakheyeva
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Evgeny Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Nadezda Cherdyntseva
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
- Tomsk State University, 36 Lenin Ave., Tomsk, 634050, Russia
| |
Collapse
|
15
|
Focaccetti C, Nardozi D, Benvenuto M, Lucarini V, Angiolini V, Carrano R, Scimeca M, Servadei F, Mauriello A, Mancini P, Besharat ZM, Milella M, Migliaccio S, Ferretti E, Cifaldi L, Masuelli L, Palumbo C, Bei R. Bisphenol-A in Drinking Water Accelerates Mammary Cancerogenesis and Favors an Immunosuppressive Tumor Microenvironment in BALB- neuT Mice. Int J Mol Sci 2024; 25:6259. [PMID: 38892447 PMCID: PMC11172679 DOI: 10.3390/ijms25116259] [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: 04/12/2024] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Bisphenol-A (BPA), a synthetic compound ubiquitously present in the environment, can act as an endocrine disruptor by binding to both canonical and non-canonical estrogen receptors (ERs). Exposure to BPA has been linked to various cancers, in particular, those arising in hormone-targeted tissues such as the breast. In this study, we evaluated the effect of BPA intake through drinking water on ErbB2/neu-driven cancerogenesis in BALB-neuT mice, transgenic for a mutated ErbB2/neu receptor gene, which reproducibly develop carcinomas in all mammary glands. In this model, BPA accelerated mammary cancerogenesis with an increase in the number of tumors per mouse and a concurrent decrease in tumor-free and overall survival. As assessed by immunohistochemistry, BALB-neuT tumors were ER-negative but expressed high levels of the alternative estrogen receptor GPR30, regardless of BPA exposure. On the other hand, BPA exposure resulted in a marked upregulation of progesterone receptors in preinvasive tumors and of Ki67, CD31, and phosphorylated Akt in invasive tumors. Moreover, based on several infiltration markers of immune cells, BPA favored an immunosuppressive tumor microenvironment. Finally, in vitro cell survival studies performed on a cell line established from a BALB-neuT breast carcinoma confirmed that BPA's impact on cancer progression can be particularly relevant after chronic, low-dose exposure.
Collapse
MESH Headings
- Animals
- Benzhydryl Compounds
- Phenols
- Tumor Microenvironment/drug effects
- Female
- Mice
- Mice, Inbred BALB C
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Drinking Water
- Mammary Neoplasms, Experimental/chemically induced
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/metabolism
- Mice, Transgenic
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/genetics
- Carcinogenesis/chemically induced
- Carcinogenesis/drug effects
- Endocrine Disruptors/toxicity
Collapse
Affiliation(s)
- Chiara Focaccetti
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (M.B.); (R.C.); (L.C.); (C.P.)
| | - Daniela Nardozi
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (D.N.); (V.L.); (V.A.); (P.M.); (Z.M.B.); (S.M.); (E.F.); (L.M.)
| | - Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (M.B.); (R.C.); (L.C.); (C.P.)
| | - Valeria Lucarini
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (D.N.); (V.L.); (V.A.); (P.M.); (Z.M.B.); (S.M.); (E.F.); (L.M.)
| | - Valentina Angiolini
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (D.N.); (V.L.); (V.A.); (P.M.); (Z.M.B.); (S.M.); (E.F.); (L.M.)
| | - Raffaele Carrano
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (M.B.); (R.C.); (L.C.); (C.P.)
| | - Manuel Scimeca
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.S.); (F.S.); (A.M.)
| | - Francesca Servadei
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.S.); (F.S.); (A.M.)
| | - Alessandro Mauriello
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.S.); (F.S.); (A.M.)
| | - Patrizia Mancini
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (D.N.); (V.L.); (V.A.); (P.M.); (Z.M.B.); (S.M.); (E.F.); (L.M.)
| | - Zein Mersini Besharat
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (D.N.); (V.L.); (V.A.); (P.M.); (Z.M.B.); (S.M.); (E.F.); (L.M.)
| | - Michele Milella
- Department of Oncology, University of Verona, 37134 Verona, Italy;
| | - Silvia Migliaccio
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (D.N.); (V.L.); (V.A.); (P.M.); (Z.M.B.); (S.M.); (E.F.); (L.M.)
| | - Elisabetta Ferretti
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (D.N.); (V.L.); (V.A.); (P.M.); (Z.M.B.); (S.M.); (E.F.); (L.M.)
| | - Loredana Cifaldi
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (M.B.); (R.C.); (L.C.); (C.P.)
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (D.N.); (V.L.); (V.A.); (P.M.); (Z.M.B.); (S.M.); (E.F.); (L.M.)
| | - Camilla Palumbo
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (M.B.); (R.C.); (L.C.); (C.P.)
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (M.B.); (R.C.); (L.C.); (C.P.)
| |
Collapse
|
16
|
Chauhan SK, Dunn C, Andresen NK, Røssevold AH, Skorstad G, Sike A, Gilje B, Raj SX, Huse K, Naume B, Kyte JA. Peripheral immune cells in metastatic breast cancer patients display a systemic immunosuppressed signature consistent with chronic inflammation. NPJ Breast Cancer 2024; 10:30. [PMID: 38653982 DOI: 10.1038/s41523-024-00638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 04/13/2024] [Indexed: 04/25/2024] Open
Abstract
Immunotherapies blocking the PD-1/PD-L1 checkpoint show some efficacy in metastatic breast cancer (mBC) but are often hindered by immunosuppressive mechanisms. Understanding these mechanisms is crucial for personalized treatments, with peripheral blood monitoring representing a practical alternative to repeated biopsies. In the present study, we performed a comprehensive mass cytometry analysis of peripheral blood immune cells in 104 patients with HER2 negative mBC and 20 healthy donors (HD). We found that mBC patients had significantly elevated monocyte levels and reduced levels of CD4+ T cells and plasmacytoid dendritic cells, when compared to HD. Furthermore, mBC patients had more effector T cells and regulatory T cells, increased expression of immune checkpoints and other activation/exhaustion markers, and a shift to a Th2/Th17 phenotype. Furthermore, T-cell phenotypes identified by mass cytometry correlated with functionality as assessed by IFN-γ production. Additional analysis indicated that previous chemotherapy and CDK4/6 inhibition impacted the numbers and phenotype of immune cells. From 63 of the patients, fresh tumor samples were analyzed by flow cytometry. Paired PBMC-tumor analysis showed moderate correlations between peripheral CD4+ T and NK cells with their counterparts in tumors. Further, a CD4+ T cell cluster in PBMCs, that co-expressed multiple checkpoint receptors, was negatively associated with CD4+ T cell tumor infiltration. In conclusion, the identified systemic immune signatures indicate an immune-suppressed environment in mBC patients who had progressed/relapsed on standard treatments, and is consistent with ongoing chronic inflammation. These activated immuno-suppressive mechanisms may be investigated as therapeutic targets, and for use as biomarkers of response or treatment resistance.
Collapse
Affiliation(s)
- Sudhir Kumar Chauhan
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Claire Dunn
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Nikolai Kragøe Andresen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Hagen Røssevold
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gjertrud Skorstad
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Adam Sike
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bjørnar Gilje
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Sunil Xavier Raj
- Department of Oncology, St Olav University Hospital, Trondheim, Norway
| | - Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bjørn Naume
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Jon Amund Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway.
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway.
| |
Collapse
|
17
|
Wang Y, Wang J, Jiang J, Zhang W, Sun L, Ge Q, Li C, Li X, Li X, Shi S. Identification of cuproptosis-related miRNAs in triple-negative breast cancer and analysis of the miRNA-mRNA regulatory network. Heliyon 2024; 10:e28242. [PMID: 38601669 PMCID: PMC11004712 DOI: 10.1016/j.heliyon.2024.e28242] [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: 06/13/2023] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024] Open
Abstract
Introduction The close association between cuproptosis and tumor immunity in triple-negative breast cancer (TNBC) allows its monitoring for predicting the prognosis of patients with TNBC. Nevertheless, the biological function and prognostic value of cuproptosis-related miRNAs and their target genes have not been reported. Purpose To construct the miRNA and mRNA-based risk models associated with cuproptosis for patients with TNBC. Methods Comparison of expression levels for genes associated with cuproptosis was executed between patients in the normal individuals and the TCGA-TNBC cohort. Conducting differential analysis resulted in the identification of differentially expressed miRNA (DE-miRNAs) and differentially expressed genes (DEGs) between the TNBC and Control samples. Screening for prognostic miRNAs and biomarkers involved employing univariate Cox analysis and least absolute shrinkage and selection operator regression analyses. These methods were utilized to construct risk models aimed at predicting the survival of patients with TNBC. Based on the median value of risk scores, patients were then stratified into low- and high-risk groups. Functional enrichment analysis was employed to explore the potential function and pathways of prognostic genes. Additionally, independent prognostic analysis was performed through univariate and multivariate Cox regression. Immune infiltration analysis was performed to examine disparities in the infiltration of immune cells between the two risk groups. Finally, the prognostic gene expression was mined in key cell types of TNBC. Results We obtained 5213 DEGs and 204 DE-miRNAs related to cuproptosis between TNBC and Control samples. Five prognostic miRNAs (miR-203a-3p, miR-1277-3p, miR-135b-5p, miR-200c-3p, and miR-592) and three biomarkers (DENND5B, IGF1R, and MEF2C) were closely associated with TNBC. Significant differences in the functions of prognostic genes between the two risk groups were observed, encompassing adipogenesis, inflammatory response, and hormone metabolic process. The prognostic gene regulatory network revealed that miR200C-3p regulated ZFPM2 and CFL2, and miR-1277-3p regulated BMP2 and RORA. A nomogram was created based on riskScore, cancer status, and pathologic stage to predict 1/3/5-year survival of patients with TNBC. Immune infiltration analysis indicated that the immune microenvironment may be associated with the progression of TNBC. Interestingly, prognostic genes exhibited higher expression levels in T cells, fibroblasts, endothelial cells, and monocytes compared to other cells. Conclusions Five prognostic miRNA (miR-203a-3p, miR-1277-3p, miR-135b-5p, miR-200c-3p, and miR-592) and three biomarkers (DENND5B, IGF1R, and MEF2C) were significantly associated with TNBC, it provides new therapeutic targets for the treatment and prognosis of TNBC.
Collapse
Affiliation(s)
- Yitao Wang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jundan Wang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jing Jiang
- Department of Breast Surgery, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Wei Zhang
- Department of Breast Surgery, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Long Sun
- Department of Breast Surgery, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Qidong Ge
- Department of Breast Surgery, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Chao Li
- Department of Breast Surgery, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Xinlin Li
- Department of Breast Surgery, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Xujun Li
- Department of Breast Surgery, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Shenghong Shi
- Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Oncology, Ningbo No.2 Hospital, Ningbo, 315010, China
- Department of Breast Surgery, Ningbo No.2 Hospital, Ningbo, 315010, China
| |
Collapse
|
18
|
Wang B, Zhou B, Chen J, Sun X, Yang W, Yang T, Yu H, Chen P, Chen K, Huang X, Fan X, He W, Huang J, Lin T. Type III interferon inhibits bladder cancer progression by reprogramming macrophage-mediated phagocytosis and orchestrating effective immune responses. J Immunother Cancer 2024; 12:e007808. [PMID: 38589249 PMCID: PMC11015199 DOI: 10.1136/jitc-2023-007808] [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] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Interferons (IFNs) are essential for activating an effective immune response and play a central role in immunotherapy-mediated immune cell reactivation for tumor regression. Type III IFN (λ), related to type I IFN (α), plays a crucial role in infections, autoimmunity, and cancer. However, the direct effects of IFN-λ on the tumor immune microenvironment have not been thoroughly investigated. METHODS We used mouse MB49 bladder tumor models, constructed a retroviral vector expressing mouse IFN-λ3, and transduced tumor cells to evaluate the antitumor action of IFN-λ3 in immune-proficient tumors and T cell-deficient tumors. Furthermore, human bladder cancer samples (cohort 1, n=15) were used for immunohistochemistry and multiplex immunoflurescence analysis to assess the expression pattern of IFN-λ3 in human bladder cancer and correlate it with immune cells' infiltration. Immunohistochemistry analysis was performed in neoadjuvant immunotherapy cohort (cohort 2, n=20) to assess the correlation between IFN-λ3 expression and the pathological complete response rate. RESULTS In immune-proficient tumors, ectopic Ifnl3 expression in tumor cells significantly increased the infiltration of cytotoxic CD8+ T cells, Th1 cells, natural killer cells, proinflammatory macrophages, and dendritic cells, but reduced neutrophil infiltration. Transcriptomic analyses revealed significant upregulation of many genes associated with effective immune response, including lymphocyte recruitment, activation, and phagocytosis, consistent with increased antitumor immune infiltrates and tumor inhibition. Furthermore, IFN-λ3 activity sensitized immune-proficient tumors to anti-PD-1/PD-L1 blockade. In T cell-deficient tumors, increased Ly6G-Ly6C+I-A/I-E+ macrophages still enhanced tumor cell phagocytosis in Ifnl3 overexpressing tumors. IFN-λ3 is expressed by tumor and stromal cells in human bladder cancer, and high IFN-λ3 expression was positively associated with effector immune infiltrates and the efficacy of immune checkpoint blockade therapy. CONCLUSIONS Our study indicated that IFN-λ3 enables macrophage-mediated phagocytosis and antitumor immune responses and suggests a rationale for using Type III IFN as a predictive biomarker and potential immunotherapeutic candidate for bladder cancer.
Collapse
Affiliation(s)
- Bo Wang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Bingkun Zhou
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Junyu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Xi Sun
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Wenjuan Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
- Department of Hematology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Tenghao Yang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Hao Yu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Peng Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Ke Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Xiaodong Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Xinxiang Fan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Wang He
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, China
| |
Collapse
|
19
|
Ramos‐Martinez E, García‐Vazquez FJ, Falfán‐Valencia R, Rojas‐Serrano J, Alfaro‐Cruz A, Pérez‐Villaseñor M, Aristi‐Urista G, Pérez‐Hernández J, López‐Vancell R, Velasco‐Medina A, Velázquez‐Sámano G. The type 2 inflammatory response favors recognition of tumor antigens by IgE in breast cancer. Cancer Rep (Hoboken) 2024; 7:e2002. [PMID: 38389406 PMCID: PMC10884619 DOI: 10.1002/cnr2.2002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/30/2023] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Several studies describe an inverse statistical relationship between the presence of an allergy and development of cancer. However, the immunological mechanism involved in the relationship between these two degenerative diseases has not been explored. AIMS The main objective of this study was to explore the possibility that the lymphocyte T helper (Th) 2 response, a characteristic of allergy, induces recognition of tumor antigens. METHODS AND RESULTS Patients with a clinical diagnosis of breast ductal carcinoma were included. Histopathological markers related to proliferation of tumor cells were determined (Her-2-neu, Ki-67, estrogen receptor, and progesterone receptor). IHC was performed using IgE antibodies purified from an allergy patient and from each biopsy donor patient. Serum concentrations of cytokines representative of Th1 and Th2 inflammatory responses were determined. A total of 14 patients with a confirmed diagnosis of breast ductal carcinoma were included. IHC performed on biopsies showed a weak response when using purified IgE antibodies from an allergy patient; however, IHC using the IgE of each patient as the primary antibody showed an intense and highly specific signal. Serum concentrations of cytokines of the Th2 response, that is, IL-4 (130.5 pg/mL (116-135 pg/mL)), IL-5 (202 pg/mL (191-213 pg/mL)), and IL-13 (105.5 pg/mL (98-117 pg/mL)), were significantly higher than those of the Th1 response, that is, IL-6 (86 pg/mL (79-90 pg/mL)) and INF-γ (93 pg/mL (79-99 pg/mL)). CONCLUSION Purified IgE antibodies specifically recognize tumor cells in breast ductal carcinoma.
Collapse
Affiliation(s)
- Espiridión Ramos‐Martinez
- Unidad de Medicina Experimental, Facultad de MedicinaUniversidad Nacional Autónoma de MéxicoMexico cityMexico
| | - Francisco Javier García‐Vazquez
- Departamento de Análisis Clínicos y Estudios EspecialesInstituto Nacional de Pediatría, Laboratorio de Inmunogenética MolecularMexico cityMexico
| | - Ramcés Falfán‐Valencia
- HLA LaboratoryInstituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico cityMexico
| | - Jorge Rojas‐Serrano
- Unidad de Enfermedades del Intersticio Pulmonar y Reumatología, Instituto Nacional de Enfermedades Respiratorias, “Ismael Cosío Villegas”Mexico cityMexico
| | - Ana Alfaro‐Cruz
- Patología Quirúrgica, Servicio de Anatomía Patológica, Hospital General de México, “Dr. Eduardo Liceaga”Mexico CityMexico
| | | | - Gerardo Aristi‐Urista
- Patología Quirúrgica, Servicio de Anatomía Patológica, Hospital General de México, “Dr. Eduardo Liceaga”Mexico CityMexico
| | - Jesús Pérez‐Hernández
- Unidad de Medicina Experimental, Facultad de MedicinaUniversidad Nacional Autónoma de MéxicoMexico cityMexico
| | - Rosario López‐Vancell
- Unidad de Medicina Experimental, Facultad de MedicinaUniversidad Nacional Autónoma de MéxicoMexico cityMexico
| | - Andrea Velasco‐Medina
- Servicio de Alergia e Inmunología Clínica, Hospital General de México, “Dr. Eduardo Liceaga”Mexico CityMexico
| | - Guillermo Velázquez‐Sámano
- Servicio de Alergia e Inmunología Clínica, Hospital General de México, “Dr. Eduardo Liceaga”Mexico CityMexico
| |
Collapse
|
20
|
Yao P, Liang S, Liu Z, Xu C. A review of natural products targeting tumor immune microenvironments for the treatment of lung cancer. Front Immunol 2024; 15:1343316. [PMID: 38361933 PMCID: PMC10867126 DOI: 10.3389/fimmu.2024.1343316] [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: 11/23/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
Lung cancer (LC) produces some of the most malignant tumors in the world, with high morbidity and mortality. Tumor immune microenvironment (TIME), a component of the tumor microenvironment (TME), are critical in tumor development, immune escape, and drug resistance. The TIME is composed of various immune cells, immune cytokines, etc, which are important biological characteristics and determinants of tumor progression and outcomes. In this paper, we reviewed the recently published literature and discussed the potential uses of natural products in regulating TIME. We observed that a total of 37 natural compounds have been reported to exert anti-cancer effects by targeting the TIME. In different classes of natural products, terpenoids are the most frequently mentioned compounds. TAMs are one of the most investigated immune cells about therapies with natural products in TIME, with 9 natural products acting through it. 17 natural products exhibit anti-cancer properties in LC by modulating PD-1 and PD-L1 protein activity. These natural products have been extensively evaluated in animal and cellular LC models, but their clinical trials in LC patients are lacking. Based on the current review, we have revealed that the mechanisms of LC can be treated with natural products through TIME intervention, resulting in a new perspective and potential therapeutic drugs.
Collapse
Affiliation(s)
- Pengyu Yao
- Department of Traditional Chinese Medicine, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Su Liang
- Department of Traditional Chinese Medicine, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhenying Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cuiping Xu
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan, China
| |
Collapse
|
21
|
Lu H, Yu X, Hou L, Zhang Y, Li L, Qiao X, Cheng H, Du L, Chen J, Zheng Q, Hou J. Analysis of CVC1302-Mediated Enhancement of Monocyte Recruitment in Inducing Immune Responses. Vaccines (Basel) 2024; 12:86. [PMID: 38250899 PMCID: PMC10820601 DOI: 10.3390/vaccines12010086] [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: 11/30/2023] [Revised: 12/25/2023] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
Monocytes (Mos) are believed to play important roles during the generation of immune response. In our previous study, CVC1302, a complex of PRRs agonists, was demonstrated to recruit Mo into lymph nodes (LNs) in order to present antigen and secret chemokines (CXCL9 and CXCL10), which attracted antigen-specific CD4+ T cells. As it is known that Mos in mice are divided into two main Mo subsets (Ly6C+ Mo and Ly6C- Mo), we aimed to clarify the CVC1302-recruiting Mo subset and functions in the establishment of immunity. In this study, we found that CVC1302 attracted both Ly6C+ Mo and Ly6C- Mo into draining LNs, which infiltrated from different origins, injection muscles and high endothelial venule (HEV), respectively. We also found that the numbers of OVA+ Ly6C+ Mo in the draining LNs were significantly higher compared with OVA+ Ly6C- Mo. However, the levels of CXCL9 and CXCL10 produced by Ly6C- Mo were significantly higher than Ly6C+ Mo, which plays important roles in attracting antigen-specific CD4+ T cells. Under the analysis of their functions in initiating immune responses, we found that the ability of the Ly6C+ monocyte was mainly capturing and presenting antigens, otherwise; the ability of the Ly6C- monocyte was mainly secreting CXCL9 and CXCL10, which attracted antigen-specific CD4+ T cells through CXCR3. These results will provide new insights into the development of new immunopotentiators and vaccines.
Collapse
Affiliation(s)
- Haiyan Lu
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoming Yu
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
| | - Liting Hou
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
| | - Yuanpeng Zhang
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
| | - Lan Li
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
| | - Xuwen Qiao
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
| | - Haiwei Cheng
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
| | - Luping Du
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
| | - Jin Chen
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
| | - Qisheng Zheng
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
| | - Jibo Hou
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Science, Nanjing 210014, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
- Guo Tai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 210014, China
| |
Collapse
|
22
|
Yu H, Liu J. Identification of breast cancer subgroups and immune characterization based on glutamine metabolism-related genes. BMC Med Genomics 2024; 17:17. [PMID: 38200578 PMCID: PMC10782609 DOI: 10.1186/s12920-023-01792-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Immunotherapy is a promising treatment for breast cancer (BC). However, due to individual differences and tumor heterogeneity, immunotherapy is only applicable to some BC patients. Glutamine metabolism plays a role in inhibiting immunotherapy, but its role in BC is limitedly studied. Therefore, we aimed to identify different BC subgroups based on glutamine metabolism and characterize the features of different subgroups to provide guidance for personalized immunotherapy for BC patients. Using unsupervised clustering analysis, we classified BC patients in The Cancer Genome Atlas (TCGA) with glutamine metabolism-related genes and obtained low-risk (LR) and high-risk (HR) subgroups. Survival analysis revealed that prognosis of LR subgroup was notably better than HR subgroup. Through ssGSEA and CIBERSORT methods, we disclosed that infiltration levels of B cells, Mast cells, T helper cells, and Th2 cells, and Type II IFN Response immune function were notably higher in LR subgroup than in HR subgroup. The Wilcox algorithm comparison denoted that DEPTH of LR subgroup was significantly lower than HR subgroup. The TIDE of LR subgroup was significantly higher than HR subgroup. Functional annotation of differentially expressed genes revealed that channel activity and the Estrogen signaling pathway may be related to BC prognosis. Ten hub genes were selected between the subgroups through the STRING database and Cytoscape, and their correlation with drugs was predicted on the CellMiner website. This study analyzed the immune characteristics of BC subgroups based on glutamine metabolism and provided reference for prognosis prediction and personalized immunotherapy.
Collapse
Affiliation(s)
- Hongjing Yu
- Department of Oncology, Jiande Branch, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Junchen Liu
- Department of Pharmacy, Jiande Branch, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
23
|
Shang Q, Yu X, Sun Q, Li H, Sun C, Liu L. Polysaccharides regulate Th1/Th2 balance: A new strategy for tumor immunotherapy. Biomed Pharmacother 2024; 170:115976. [PMID: 38043444 DOI: 10.1016/j.biopha.2023.115976] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/20/2023] [Accepted: 11/29/2023] [Indexed: 12/05/2023] Open
Abstract
T helper (Th) cells have received extensive attention owing to their indispensable roles in anti-tumor immune responses. Th1 and Th2 cells are two key subsets of Th cells that exist in relative equilibrium through the secretion of cytokines that suppress their respective immune response. When the type of cytokine in the tumor microenvironment is altered, this equilibrium may be disrupted, leading to a shift from Th1 to Th2 immune response. Th1/Th2 imbalance is one of the decisive factors in the development of malignant tumors. Therefore, focusing on the balance of Th1/Th2 anti-tumor immune responses may enable future breakthroughs in cancer immunotherapy. Polysaccharides can regulate the imbalance between Th1 and Th2 cells and their characteristic cytokine profiles, thereby improving the tumor immune microenvironment. To our knowledge, this study is the most comprehensive assessment of the regulation of the tumor Th1/Th2 balance by polysaccharides. Herein, we systematically summarized the intrinsic molecular mechanisms of polysaccharides in the regulation of Th1 and Th2 cells to provide a new perspective and potential target drugs for improved anti-tumor immunity and delayed tumor progression.
Collapse
Affiliation(s)
- Qihang Shang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiaoyun Yu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Qi Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China; Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| |
Collapse
|
24
|
Liu Y, Liu S, Yan L, Zhang Q, Liu W, Huang X, Liu S. Contribution of m5C RNA Modification-Related Genes to Prognosis and Immunotherapy Prediction in Patients with Ovarian Cancer. Mediators Inflamm 2023; 2023:1400267. [PMID: 38022687 PMCID: PMC10661868 DOI: 10.1155/2023/1400267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/03/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Background 5-Methylcytosine (m5C) RNA modification is closely implicated in the occurrence of a variety of cancers. Here, we established a novel prognostic signature for ovarian cancer (OC) patients based on m5C RNA modification-related genes and explored the correlation between these genes with the tumor immune microenvironment. Methods Methylated-RNA immunoprecipitation sequencing helped us to identify candidate genes related to m5C RNA modification at first. Based on TCGA database, we screened the differentially expressed candidate genes related to the prognosis and constructed a prognostic model using LASSO Cox regression analyses. Notably, the accuracy of the model was evaluated by Kaplan-Meier analysis and receiver operator characteristic curves. Independent prognostic risk factors were investigated by Cox proportional hazard model. Furthermore, we also analyzed the biological functions and pathways involved in the signature. Finally, the immune response of the model was visualized in great detail. Results Totally, 2,493 candidate genes proved to be involved in m5C modification of RNA for OC. We developed a signature with prognostic value consisting of six m5C RNA modification-related genes. Specially, samples have been split into two cohorts with low- and high-risk scores according to the model, in which the low-risk OC patients exhibited dramatically better overall survival time than those with high-risk scores. Besides, not only was this model a prognostic factor independent of other clinical characteristics but it predicted the intensity of the immune response in OC. Significantly, the accuracy and availability of the signature were verified by ICGC database. Conclusions Our study bridged the gap between m5C RNA modification and the prognosis of OC and was expected to provide an effective breakthrough for immunotherapy in OC patients.
Collapse
Affiliation(s)
- Yibin Liu
- Department of Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei 050011, China
| | - Shouze Liu
- Department of Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei 050011, China
- Department of Gynecology III, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| | - Lu Yan
- Department of Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei 050011, China
| | - Qianqian Zhang
- Department of Gynecology and Obstetrics, Beijing Tsinghua Changgung Hospital, Beijing 102218, China
| | - Wenhua Liu
- Department of Pain, Cangzhou Hospital of Integrated TCM-WM Hebei, Cangzhou, Hebei 061001, China
| | - Xianghua Huang
- Department of Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei 050011, China
| | - Shikai Liu
- Department of Gynecology III, Cangzhou Central Hospital, Cangzhou, Hebei 061000, China
| |
Collapse
|
25
|
Azin M, Ngo KH, Hojanazarova J, Demehri S. Topical Calcipotriol Plus Imiquimod Immunotherapy for Nonkeratinocyte Skin Cancers. JID INNOVATIONS 2023; 3:100221. [PMID: 37731472 PMCID: PMC10507651 DOI: 10.1016/j.xjidi.2023.100221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 09/22/2023] Open
Abstract
Nonkeratinocyte cutaneous malignancies, including breast cancer cutaneous metastasis and melanoma in situ, are often poor surgical candidates. Imiquimod (IMQ), a toll-like receptor 7 agonist that activates innate immunity in the skin, is used to treat these cutaneous malignancies. However, IMQ's modest effect on the activation of adaptive immunity limits its efficacy as a monotherapy. In this study, we demonstrate that topical TSLP cytokine inducers-calcipotriol and retinoic acid-synergize with IMQ to activate CD4+ T-cell immunity against nonkeratinocyte cutaneous malignancies. Topical calcipotriol plus IMQ treatment reduced breast tumor growth compared with calcipotriol or IMQ alone (P < 0.0001). Calcipotriol plus IMQ-mediated tumor suppression was associated with significant infiltration of CD4+ effector T cells in the tumor microenvironment. Notably, topical calcipotriol plus IMQ immunotherapy enabled immune checkpoint blockade therapy to effectively control immunologically cold breast tumors, which was associated with induction of CD4+ T-cell immunity. Topical treatment with calcipotriol plus IMQ and retinoic acid plus IMQ also blocked subcutaneous melanoma growth. These findings highlight the synergistic effect of topical TSLP induction in combination with innate immune cell activation as an effective immunotherapy for malignancies affecting the skin.
Collapse
Affiliation(s)
- Marjan Azin
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kenneth H. Ngo
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jennet Hojanazarova
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shadmehr Demehri
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Li J, Liu Y, Zeng W, Wu Y, Ao W, Yuan X, Zhou C. The Relationship Between the Expression of circFAT1 and Immune Cell in Patients with Non-Small Cell Lung Cancer. Int J Gen Med 2023; 16:4943-4951. [PMID: 37928955 PMCID: PMC10625319 DOI: 10.2147/ijgm.s434065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023] Open
Abstract
Objective To analyze the correlation between the expression of circFAT1 in serum and immune cells in patients with non-small cell lung cancer (NSCLC). Methods A total of 96 patients with NSCLC admitted to our hospital from November 2019 to November 2022 were regarded as the study subjects. In the meantime, 96 volunteers who had physical examination in our hospital were regarded as the control group. The expression level of circFAT1 in serum was detected by real-time fluorescence quantitative PCR. NSCLC cancer tissue (NSCLC group) and paracancerous tissue (tissue ≥ 2cm away from the focus) (paracancerous group) were collected during the operation, the expression of CD4+, CD8+ and Foxp3+ in tissues was determined by immunohistochemistry; the expression level of circFAT1 mRNA in NSCLC tissue was analyzed using the Ualcan database. Spearman correlation was applied to analyze the correlation between the expression of circFAT1 and immune cells (CD4+, Foxp3+, CD8+). Results The level of circFAT1 in NSCLC tissue was higher than that in normal tissue (P < 0.05). Compared with the control group, the expression level of circFAT1 in serum of NSCLC group was obviously higher (P < 0.05). The expression level of circFAT1 was related to lymph node metastasis, TNM stage and differentiation (P < 0.05). Compared with the paracancerous group, the positive expression rate of CD8+ in NSCLC group was obviously lower, and the positive expression rates of CD4+ and Foxp3+ were obviously higher (P < 0.05). The expression of CD4+, Foxp3+ and CD8+ in NSCLC patients' cancer tissue was related to lymph node metastasis, TNM stage and differentiation degree (P < 0.05). Spearman correlation analysis showed that circFAT1 was positively correlated with the expression of CD4+ and Foxp3+ and negatively correlated with the expression of CD8+ (P < 0.05). Conclusion CircFAT1 is highly expressed in the serum of NSCLC patients and is closely related to immune cells.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Oncology, Yueyang People’s Hospital, Yueyang City, Hunan Province, 414000, People’s Republic of China
| | - Yabing Liu
- Department of Oncology, Yueyang People’s Hospital, Yueyang City, Hunan Province, 414000, People’s Republic of China
| | - Wenxuan Zeng
- Department of Cardiovascular, Yueyang Central Hospital, Yueyang City, Hunan Province, 414000, People’s Republic of China
| | - Yanrun Wu
- Department of Ultrasonic, Yueyang Central Hospital, Yueyang City, Hunan Province, 414000, People’s Republic of China
| | - Wei Ao
- Department of Cardiovascular, Yueyang People’s Hospital, Yueyang City, Hunan Province, 414000, People’s Republic of China
| | - Xiwei Yuan
- Department of Oncology, Yueyang People’s Hospital, Yueyang City, Hunan Province, 414000, People’s Republic of China
| | - Chuanyi Zhou
- Department of Oncology, Yueyang People’s Hospital, Yueyang City, Hunan Province, 414000, People’s Republic of China
| |
Collapse
|
27
|
Zareinejad M, Mehdipour F, Roshan-Zamir M, Faghih Z, Ghaderi A. Dual Functions of T Lymphocytes in Breast Carcinoma: From Immune Protection to Orchestrating Tumor Progression and Metastasis. Cancers (Basel) 2023; 15:4771. [PMID: 37835465 PMCID: PMC10571747 DOI: 10.3390/cancers15194771] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer (BC) is the most common cancer type in women and the second leading cause of death. Despite recent advances, the mortality rate of BC is still high, highlighting a need to develop new treatment strategies including the modulation of the immune system and immunotherapies. In this regard, understanding the complex function of the involved immune cells and their crosstalk with tumor cells is of great importance. T-cells are recognized as the most important cells in the tumor microenvironment and are divided into several subtypes including helper, cytotoxic, and regulatory T-cells according to their transcription factors, markers, and functions. This article attempts to provide a comprehensive review of the role of T-cell subsets in the prognosis and treatment of patients with BC, and crosstalk between tumor cells and T-cells. The literature overwhelmingly contains controversial findings mainly due to the plasticity of T-cell subsets within the inflammatory conditions and the use of different panels for their phenotyping. However, investigating the role of T-cells in BC immunity depends on a variety of factors including tumor types or subtypes, the stage of the disease, the localization of the cells in the tumor tissue and the presence of different cells or cytokines.
Collapse
Affiliation(s)
| | | | | | - Zahra Faghih
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-45505, Iran; (M.Z.); (F.M.); (M.R.-Z.)
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-45505, Iran; (M.Z.); (F.M.); (M.R.-Z.)
| |
Collapse
|
28
|
Chen L, Wang Y, Hu Q, Liu Y, Qi X, Tang Z, Hu H, Lin N, Zeng S, Yu L. Unveiling tumor immune evasion mechanisms: abnormal expression of transporters on immune cells in the tumor microenvironment. Front Immunol 2023; 14:1225948. [PMID: 37545500 PMCID: PMC10401443 DOI: 10.3389/fimmu.2023.1225948] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/06/2023] [Indexed: 08/08/2023] Open
Abstract
The tumor microenvironment (TME) is a crucial driving factor for tumor progression and it can hinder the body's immune response by altering the metabolic activity of immune cells. Both tumor and immune cells maintain their proliferative characteristics and physiological functions through transporter-mediated regulation of nutrient acquisition and metabolite efflux. Transporters also play an important role in modulating immune responses in the TME. In this review, we outline the metabolic characteristics of the TME and systematically elaborate on the effects of abundant metabolites on immune cell function and transporter expression. We also discuss the mechanism of tumor immune escape due to transporter dysfunction. Finally, we introduce some transporter-targeted antitumor therapeutic strategies, with the aim of providing new insights into the development of antitumor drugs and rational drug usage for clinical cancer therapy.
Collapse
Affiliation(s)
- Lu Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang, Department of Clinical Pharmacy, Affiliated Hangzhou First People’s Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, China
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuchen Wang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qingqing Hu
- The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Jinhua, China
| | - Yuxi Liu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Haihong Hu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang, Department of Clinical Pharmacy, Affiliated Hangzhou First People’s Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Lushan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of Pharmacy, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, China
- Department of Pharmacy, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 295] [Impact Index Per Article: 147.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
30
|
Li F, Song X, Zhou X, Chen L, Zheng J. Emodin attenuates high lipid-induced liver metastasis through the AKT and ERK pathways in vitro in breast cancer cells and in a mouse xenograft model. Heliyon 2023; 9:e17052. [PMID: 37484373 PMCID: PMC10361095 DOI: 10.1016/j.heliyon.2023.e17052] [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: 06/28/2022] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 07/25/2023] Open
Abstract
Emodin, a natural anthraquinone derivative, can inhibit lipid synthesis and breast cancer cell proliferation. We previously found that emodin decreased breast cancer liver metastasis via epithelial-to-mesenchymal transition (EMT) inhibition. However, the mechanism through which emodin affects breast cancer liver metastasis in high-fat diet-induced obese and hyperlipidemic mice has not been elucidated. Bioinformatics analysis was used to reveal the potential targets and pathways of emodin. The mouse model of liver metastasis was established by injecting breast cancer cells into the left ventricle in high-fat diet-induced obese mice. The effect of emodin on inhibiting liver metastasis of breast cancer was evaluated by animal experiments. The mechanisms through which emodin inhibits liver metastasis of breast cancer were studied by cell and molecular biological methods. Emodin reduced lipid synthesis by inhibiting the expression of triglyceride (TG) synthesis-related genes, such as fatty acid synthase (Fasn), glycerol-3-phosphate acyltransferase 1 (Gpat1), and stearoyl-CoA desaturase (Scd1), and ultimately reduced liver metastasis in breast cancer. In addition, emodin inhibited breast cancer cell proliferation and invasion through the serine/threonine kinase (AKT) signaling and extracellular-regulated protein kinase (ERK) pathways by interacting with CSNK2A1, ESR1, ESR2, PIM1 and PTP4A3. Our results indicate that emodin may have therapeutic potential in the prevention or treatment of breast cancer liver metastasis.
Collapse
|
31
|
Yang Y, Liu H, Chen Y, Xiao N, Zheng Z, Liu H, Wan J. Liquid biopsy on the horizon in immunotherapy of non-small cell lung cancer: current status, challenges, and perspectives. Cell Death Dis 2023; 14:230. [PMID: 37002211 PMCID: PMC10066332 DOI: 10.1038/s41419-023-05757-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 04/03/2023]
Abstract
Non-small cell lung cancer (NSCLC) is one of the most threatening malignancies to human health and life. In most cases, patients with NSCLC are already at an advanced stage when they are diagnosed. In recent years, lung cancer has made great progress in precision therapy, but the efficacy of immunotherapy is unstable, and its response rate varies from patient to patient. Several biomarkers have been proposed to predict the outcomes of immunotherapy, such as programmed cell death-ligand 1 (PD-L1) expression and tumor mutational burden (TMB). Nevertheless, the detection assays are invasive and demanding on tumor tissue. To effectively predict the outcomes of immunotherapy, novel biomarkers are needed to improve the performance of conventional biomarkers. Liquid biopsy is to capture and detect circulating tumor cells (CTCs), circulating tumor DNA (ctDNA) and exosomes in body fluids, such as blood, saliva, urine, pleural fluid and cerebrospinal fluid as samples from patients, so as to make analysis and diagnosis of cancer and other diseases. The application of liquid biopsy provides a new possible solution, as it has several advantages such as non-invasive, real-time dynamic monitoring, and overcoming tumor heterogeneity. Liquid biopsy has shown predictive value in immunotherapy, significantly improving the precision treatment of lung cancer patients. Herein, we review the application of liquid biopsy in predicting the outcomes of immunotherapy in NSCLC patients, and discuss the challenges and future directions in this field.
Collapse
Affiliation(s)
- Ying Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Youming Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Nan Xiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhaoyang Zheng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Hongchun Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
32
|
Arpinati L, Scherz-Shouval R. From gatekeepers to providers: regulation of immune functions by cancer-associated fibroblasts. Trends Cancer 2023; 9:421-443. [PMID: 36870916 DOI: 10.1016/j.trecan.2023.01.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 03/06/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are major protumorigenic components of the tumor microenvironment in solid cancers. CAFs are heterogeneous, consisting of multiple subsets that display diverse functions. Recently, CAFs have emerged as major promoters of immune evasion. CAFs favor T cell exclusion and exhaustion, promote recruitment of myeloid-derived suppressor cells, and induce protumoral phenotypic shifts in macrophages and neutrophils. With the growing appreciation of CAF heterogeneity came the understanding that different CAF subpopulations may be driving distinct immune-regulatory effects, interacting with different cell types, and perhaps even driving opposing effects on malignancy. In this review we discuss the current understanding of CAF-immune interactions, their effect on tumor progression and therapeutic response, and the possibility of exploiting CAF-immune interactions as potential targets for cancer therapy.
Collapse
Affiliation(s)
- Ludovica Arpinati
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
33
|
Lopes AD, Galdino NAL, Figueiredo AB, Brianese RC, Morais KLP, De Brot M, Osório CABT, Teixeira-Carvalho A, Calsavara VF, Evangelista GFB, Alves NS, Makdissi FB, Sanches SM, Cordeiro de Lima VC, Carraro DM, Gollob KJ. Systemic immune mediators reflect tumour-infiltrating lymphocyte intensity and predict therapeutic response in triple-negative breast cancer. Immunology 2023; 169:229-241. [PMID: 36703241 DOI: 10.1111/imm.13627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer (BC). Neoadjuvant chemotherapy has proven efficacy in its treatment, and a pathological complete response (pCR) to therapy is predictive of improved long-term survival. The immune response is key to successful neoadjuvant chemotherapy, as indicated by the relation between the percentage of stromal tumour-infiltrating lymphocytes (TILs) in pre-treated tumour tissue samples and the likelihood of achieving pCR. Here we studied systemic immune mediators from volunteer TNBC patients before undergoing neoadjuvant chemotherapy to determine the systemic response association with TIL intensity, treatment response and survival. Patients were classified into pCR responder or non-responder at time of surgery. We found higher levels of immune mediators before treatment began in patients that went on to be pCR responders versus non-pCR, with area under the curve (AUC) values of 0.64-0.80. We also observed a positive correlation between inflammatory systemic immune mediators and the percentage of TILs in pCR responder patients. Combining TILs and systemic immune mediator levels provided stronger AUC values (range of 0.72-0.82). Last, performing a progression-free survival analysis with several of the systemic cytokines that predict pCR, segregated the patients into long- and short-survival groups based on high and low production of the cytokines, respectively. Our study demonstrates that circulating cytokines, before treatment begins, predict pCR in TNBC patients treated with neoadjuvant chemotherapy. Moreover, they may act as a surrogate marker of high TILs or together with TILs to better predict pCR and survival.
Collapse
Affiliation(s)
- Ananda D Lopes
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Nayane A L Galdino
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Amanda B Figueiredo
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Rafael C Brianese
- Laboratory of Genomics and Molecular Biology, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Katia L P Morais
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Marina De Brot
- Department of Pathology, A.C. Camargo Cancer Center, São Paulo, Brazil
| | | | | | - Vinicius F Calsavara
- Laboratory of Epidemiology and Statistics, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Guilherme F B Evangelista
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Natalia S Alves
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | | | - Solange M Sanches
- Department of Mastology, A.C.Camargo Cancer Center, São Paulo, Brazil
| | | | - Dirce M Carraro
- Laboratory of Genomics and Molecular Biology, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,INCT-INCITO, São Paulo, Brazil
| | - Kenneth J Gollob
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,INCT-INCITO, São Paulo, Brazil.,Center for Research in Immuno-oncology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
34
|
Silva RCMC, Lopes MF, Travassos LH. Distinct T helper cell-mediated antitumor immunity: T helper 2 cells in focus. CANCER PATHOGENESIS AND THERAPY 2023; 1:76-86. [PMID: 38328613 PMCID: PMC10846313 DOI: 10.1016/j.cpt.2022.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/07/2022] [Accepted: 11/02/2022] [Indexed: 02/09/2024]
Abstract
The adaptive arm of the immune system is crucial for appropriate antitumor immune responses. It is generally accepted that clusters of differentiation 4+ (CD4+) T cells, which mediate T helper (Th) 1 immunity (type 1 immunity), are the primary Th cell subtype associated with tumor elimination. In this review, we discuss evidence showing that antitumor immunity and better prognosis can be associated with distinct Th cell subtypes in experimental mouse models and humans, with a focus on Th2 cells. The aim of this review is to provide an overview and understanding of the mechanisms associated with different tumor outcomes in the face of immune responses by focusing on the (1) site of tumor development, (2) tumor properties (i. e., tumor metabolism and cytokine receptor expression), and (3) type of immune response that the tumor initially escaped. Therefore, we discuss how low-tolerance organs, such as lungs and brains, might benefit from a less tissue-destructive immune response mediated by Th2 cells. In addition, Th2 cells antitumor effects can be independent of CD8+ T cells, which would circumvent some of the immune escape mechanisms that tumor cells possess, like low expression of major histocompatibility-I (MHC-I). Finally, this review aims to stimulate further studies on the role of Th2 cells in antitumor immunity and briefly discusses emerging treatment options.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Marcela Freitas Lopes
- Laboratory of Immunity Biology George DosReis,Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Leonardo Holanda Travassos
- Laboratory of Immunoreceptors and Signaling, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
35
|
Chen Y, Sun J, Luo Y, Liu J, Wang X, Feng R, Huang J, Du H, Li Q, Tan J, Ren G, Wang X, Li H. Pharmaceutical targeting Th2-mediated immunity enhances immunotherapy response in breast cancer. J Transl Med 2022; 20:615. [PMID: 36564797 PMCID: PMC9783715 DOI: 10.1186/s12967-022-03807-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer is a complex disease with a highly immunosuppressive tumor microenvironment, and has limited clinical response to immune checkpoint blockade (ICB) therapy. T-helper 2 (Th2) cells, an important component of the tumor microenvironment (TME), play an essential role in regulation of tumor immunity. However, the deep relationship between Th2-mediated immunity and immune evasion in breast cancer remains enigmatic. METHODS Here, we first used bioinformatics analysis to explore the correlation between Th2 infiltration and immune landscape in breast cancer. Suplatast tosilate (IPD-1151 T, IPD), an inhibitor of Th2 function, was then employed to investigate the biological effects of Th2 blockade on tumor growth and immune microenvironment in immunocompetent murine breast cancer models. The tumor microenvironment was analyzed by flow cytometry, mass cytometry, and immunofluorescence staining. Furthermore, we examined the efficacy of IPD combination with ICB treatment by evaluating TME, tumor growth and mice survival. RESULTS Our bioinformatics analysis suggested that higher infiltration of Th2 cells indicates a tumor immunosuppressive microenvironment in breast cancer. In three murine breast cancer models (EO771, 4T1 and EMT6), IPD significantly inhibited the IL-4 secretion by Th2 cells, promoted Th2 to Th1 switching, remodeled the immune landscape and inhibited tumor growth. Remarkably, CD8+ T cell infiltration and the cytotoxic activity of cytotoxic T lymphocyte (CTL) in tumor tissues were evidently enhanced after IPD treatment. Furthermore, increased effector CD4+ T cells and decreased myeloid-derived suppressor cells and M2-like macrophages were also demonstrated in IPD-treated tumors. Importantly, we found IPD reinforced the therapeutic response of ICB without increasing potential adverse effects. CONCLUSIONS Our findings demonstrate that pharmaceutical inhibition of Th2 cell function improves ICB response via remodeling immune landscape of TME, which illustrates a promising combinatorial immunotherapy.
Collapse
Affiliation(s)
- Yuru Chen
- grid.452206.70000 0004 1758 417XChongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China ,grid.452206.70000 0004 1758 417XDepartment of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Jiazheng Sun
- grid.452206.70000 0004 1758 417XChongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China ,grid.452206.70000 0004 1758 417XDepartment of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Yachan Luo
- grid.452206.70000 0004 1758 417XDepartment of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Jiazhou Liu
- grid.452206.70000 0004 1758 417XChongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China ,grid.452206.70000 0004 1758 417XDepartment of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Xiaoyu Wang
- grid.452206.70000 0004 1758 417XChongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China ,grid.452206.70000 0004 1758 417XDepartment of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Rui Feng
- grid.452206.70000 0004 1758 417XChongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China ,grid.452206.70000 0004 1758 417XDepartment of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Jing Huang
- grid.452206.70000 0004 1758 417XDepartment of Respiratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Huimin Du
- grid.452206.70000 0004 1758 417XDepartment of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Qin Li
- grid.411610.30000 0004 1764 2878Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050 China
| | - Jinxiang Tan
- grid.452206.70000 0004 1758 417XDepartment of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Guosheng Ren
- grid.452206.70000 0004 1758 417XChongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China ,grid.452206.70000 0004 1758 417XDepartment of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Xiaoyi Wang
- grid.452206.70000 0004 1758 417XDepartment of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Hongzhong Li
- grid.452206.70000 0004 1758 417XChongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China ,grid.452206.70000 0004 1758 417XDepartment of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| |
Collapse
|
36
|
Stanbery AG, Shuchi Smita, Jakob von Moltke, Tait Wojno ED, Ziegler SF. TSLP, IL-33, and IL-25: Not just for allergy and helminth infection. J Allergy Clin Immunol 2022; 150:1302-1313. [PMID: 35863509 PMCID: PMC9742339 DOI: 10.1016/j.jaci.2022.07.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/16/2022] [Accepted: 07/08/2022] [Indexed: 12/14/2022]
Abstract
The release of cytokines from epithelial and stromal cells is critical for the initiation and maintenance of tissue immunity. Three such cytokines, thymic stromal lymphopoietin, IL-33, and IL-25, are important regulators of type 2 immune responses triggered by parasitic worms and allergens. In particular, these cytokines activate group 2 innate lymphoid cells, TH2 cells, and myeloid cells, which drive hallmarks of type 2 immunity. However, emerging data indicate that these tissue-associated cytokines are not only involved in canonical type 2 responses but are also important in the context of viral infections, cancer, and even homeostasis. Here, we provide a brief review of the roles of thymic stromal lymphopoietin, IL-33, and IL-25 in diverse immune contexts, while highlighting their relative contributions in tissue-specific responses. We also emphasize a biologically motivated framework for thinking about the integration of multiple immune signals, including the 3 featured in this review.
Collapse
Affiliation(s)
| | - Shuchi Smita
- Department of Immunology, University of Washington, Seattle, Wash
| | - Jakob von Moltke
- Department of Immunology, University of Washington, Seattle, Wash
| | | | - Steven F Ziegler
- Department of Immunology, University of Washington, Seattle, Wash; Benaroya Research Institute, Seattle, Wash.
| |
Collapse
|
37
|
Boieri M, Marchese E, Pham QM, Azin M, Steidl LE, Malishkevich A, Demehri S. Thymic stromal lymphopoietin-stimulated CD4+ T cells induce senescence in advanced breast cancer. Front Cell Dev Biol 2022; 10:1002692. [PMID: 36467403 PMCID: PMC9714463 DOI: 10.3389/fcell.2022.1002692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/20/2022] [Indexed: 11/18/2022] Open
Abstract
Thymic Stromal Lymphopoietin (TSLP) plays a prominent role in inducing type 2 immune response, commonly associated with atopic diseases. TSLP-activated CD4+ T helper 2 cells block early carcinogenesis by inducing terminal differentiation in spontaneous breast and lung cancer models. However, the impact of TSLP induction on advanced cancer with altered cellular phenotypes is unclear. Using an established MMTV-PyMttg breast cancer cell line, we demonstrate that TSLP-stimulated CD4+ T cells possess an antitumor effect in advanced breast cancer. In contrast to early breast cancer suppression, the antitumor immunity mediated by TSLP-stimulated CD4+ T cells in advanced breast cancer is mediated by the induction of a senescent-like phenotype in cancer cells. Inflammatory CD4+ T cells drive breast cancer cells into senescence by releasing interferon-gamma and tumor necrosis factor-alpha, which directly bind to their receptors on cancer cells. Our findings reveal a novel mechanism of TSLP-activated CD4+ T cell immunity against advanced breast cancer, mediated by cellular senescence as a distinct effector mechanism for cancer immunotherapy.
Collapse
Affiliation(s)
- Margherita Boieri
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Emanuela Marchese
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Quan Minh Pham
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Marjan Azin
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Lauren E. Steidl
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Anna Malishkevich
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Shadmehr Demehri
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- *Correspondence: Shadmehr Demehri,
| |
Collapse
|
38
|
Crump LS, Kines KT, Richer JK, Lyons TR. Breast cancers co-opt normal mechanisms of tolerance to promote immune evasion and metastasis. Am J Physiol Cell Physiol 2022; 323:C1475-C1495. [PMID: 36189970 PMCID: PMC9662806 DOI: 10.1152/ajpcell.00189.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022]
Abstract
Normal developmental processes, such as those seen during embryonic development and postpartum mammary gland involution, can be reactivated by cancer cells to promote immune suppression, tumor growth, and metastatic spread. In mammalian embryos, paternal-derived antigens are at risk of being recognized as foreign by the maternal immune system. Suppression of the maternal immune response toward the fetus, which is mediated in part by the trophoblast, is critical to ensure embryonic survival and development. The postpartum mammary microenvironment also exhibits immunosuppressive mechanisms accompanying the massive cell death and tissue remodeling that occurs during mammary gland involution. These normal immunosuppressive mechanisms are paralleled during malignant transformation, where tumors can develop neoantigens that may be recognized as foreign by the immune system. To circumvent this, tumors can dedifferentiate and co-opt immune-suppressive mechanisms normally utilized during fetal tolerance and postpartum mammary involution. In this review, we discuss those similarities and how they can inform our understanding of cancer progression and metastasis.
Collapse
Affiliation(s)
- Lyndsey S Crump
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kelsey T Kines
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, Aurora, Colorado
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, Aurora, Colorado
| |
Collapse
|