1
|
Niu R, Wang C, Xie Y, Li S, Zhao Q, Chang Y, Mei Z. Prognostic significance of CD8 + tumor-infiltrating lymphocytes in operable breast cancer: a meta-analysis. BMC Cancer 2025; 25:601. [PMID: 40175948 PMCID: PMC11967132 DOI: 10.1186/s12885-025-13912-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/11/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND As key mediators of antitumor immunity, CD8 + tumor-infiltrating lymphocytes present antigens and initiate robust immune responses against cancer cells. When stratified by location, CD8 + T lymphocytes were counted and classified as intratumoral, stromal, or total CD8 + tumor-infiltrating lymphocytes. Despite their crucial role, the impact, especially the specific type of CD8 + T lymphocytes on breast cancer prognosis remains controversial. This meta-analysis synthesized evidence to delineate the relationship between CD8 + tumor-infiltrating lymphocytes density of different counting methods and breast cancer patient outcomes. METHODS PubMed, Embase, and the Cochrane Library were systemically searched from inception through January 2024 for studies evaluating the prognostic significance of CD8 + tumor-infiltrating lymphocytes in breast cancer. The primary endpoint was disease-free survival (DFS), and the second endpoints were overall survival (OS), breast cancer-specific survival (BCSS), and recurrence-free survival (RFS). RESULTS Thirty-four studies encompassing 23,626 breast cancer patients were included. Pooled hazard ratios (HRs) indicated a significant association of high CD8 + TIL presence with improved DFS (HR = 0.63; 95% CI = 0.54-0.73), OS (HR = 0.72; 95% CI = 0.65-0.79), BCSS (HR = 0.67; 95% CI = 0.58-0.78), and RFS (HR = 0.53; 95% CI = 0.38-0.73). Stratification by TIL location (intratumoral [iCD8], stromal [sCD8], or total [tCD8]) did not significantly impact DFS or OS. CONCLUSION High CD8 + TIL density in breast cancer patients is correlated with a favorable prognosis, irrespective of the location of CD8 + tumor-infiltrating lymphocytes. These findings affirm the prognostic utility of CD8 + TIL assessment and may guide future immunotherapeutic strategies.
Collapse
Affiliation(s)
- Ruijie Niu
- Department of Breast Surgery, Huangpu Branch of the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Cheng Wang
- Department of Breast Surgery, Huangpu Branch of the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Yiqun Xie
- Department of Breast Surgery, Huangpu Branch of the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shuangshuang Li
- Department of Pathology, Huangpu Branch of the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Qian Zhao
- Department of Pathology, Huangpu Branch of the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Yuqing Chang
- Department of Pathology, Huangpu Branch of the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Zubing Mei
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 201203, China.
- Anorectal Disease Institute of Shuguang Hospital, Shanghai, People's Republic of China.
| |
Collapse
|
2
|
Ni X, Wang W, Sun H, An R, Lei Y, Wang CL. A new marker for predicting sentinel lymph node metastasis in early (cT1-2N0) breast cancer: Tumor-infiltrating lymphocytes (TILs). PLoS One 2025; 20:e0320487. [PMID: 40106761 PMCID: PMC11922523 DOI: 10.1371/journal.pone.0320487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs) are associated with lymph node metastasis and prognosis in breast cancer. Therefore, we explored the value of TILs in predicting sentinel lymph node metastasis (SLNM) in patients with early-stage (cT1-2N0) breast cancer and provided a new method for preoperative assessment of SLNM status. METHODS This study included 337 patients with early-stage breast cancer who underwent surgery at our hospital from January 2022 to December 2023. The expression of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), and Ki-67 in the patients was assessed using immunohistochemistry (IHC). TILs in the core needle biopsy samples were evaluated histopathologically, and patients were divided into high and low TILs groups based on the density of TILs. Statistical analysis was conducted, and a predictive model was established. RESULTS The study found that patients with high TILs had a significantly lower rate of SLNM compared to those with low TILs (P < 0.001). The cT stage and the level of TILs were identified as independent predictive factors for SLNM. The ROC curve analysis indicated that the density of TILs has good predictive efficacy for SLNM. Based on the results of the multivariate regression analysis, a nomogram predictive model for SLNM was constructed. CONCLUSIONS Our study showed that the density of TILs and cT stage are independent predictive factors for SLNM in early-stage (cT1-2N0) breast cancer, and the predictive effect of TILs density on SLNM is significant in Luminal and triple-negative breast cancers.
Collapse
Affiliation(s)
- Xihao Ni
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, People’s Republic of China,
| | - Weitao Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, People’s Republic of China,
| | - Huimin Sun
- Department of Pathology, Weifang People’s Hospital, Weifang, Shandong Province, People’s Republic of China,
| | - Ran An
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, People’s Republic of China,
| | - Ying Lei
- Department of Breast Surgery, Weifang People’s Hospital, Weifang, Shandong Province, People’s Republic of China
| | - Chang-liang Wang
- Department of Breast Surgery, Weifang People’s Hospital, Weifang, Shandong Province, People’s Republic of China
| |
Collapse
|
3
|
Kaur K, Jewett A. Distinct profiles of osteoclast and dendritic cell-mediated expansion and functional activation of NK and T cells. Cancer Immunol Immunother 2025; 74:127. [PMID: 40024920 PMCID: PMC11872835 DOI: 10.1007/s00262-025-03956-x] [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: 09/03/2024] [Accepted: 01/22/2025] [Indexed: 03/04/2025]
Abstract
Osteoclasts (OCs) and dendritic cells (DCs) induce expansion and functional activation of NK and T cells. When comparing OCs with DC-induced activation in NK cells, OCs induced significantly higher cell expansion and functional activation of NK cells as compared to DCs, either from healthy individuals or those obtained from cancer patients. However, no differences could be seen in the levels of cell expansion and functional activation in T cells activated by OCs or DCs, either from healthy individuals or those from cancer patients. OCs selectively expanded and activated CD8 + T cells, whereas DCs expanded and activated CD4 + T cells. In addition, both allogeneic and autologous OCs induced similar levels of cell expansion and functional activation of NK and T cells. Together, these findings highlighted the essential roles of OCs in expanding and activating the cytotoxic effectors of NK, and CD8 + T cells, and demonstrated several differences when compared to the effect of DCs.
Collapse
Affiliation(s)
- Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave., Los Angeles, CA, 90095, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave., Los Angeles, CA, 90095, USA.
- The Jonsson Comprehensive Cancer Center, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave., Los Angeles, CA, 90095, USA.
| |
Collapse
|
4
|
Moura T, Caramelo O, Silva I, Silva S, Gonçalo M, Portilha MA, Moreira JN, Gil AM, Laranjeira P, Paiva A. Early-Stage Luminal B-like Breast Cancer Exhibits a More Immunosuppressive Tumor Microenvironment than Luminal A-like Breast Cancer. Biomolecules 2025; 15:78. [PMID: 39858472 PMCID: PMC11763923 DOI: 10.3390/biom15010078] [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: 11/26/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Breast cancer is a heterogeneous malignant disease with a varying prognosis and is classified into four molecular subtypes. It remains one of the most prevalent cancers globally, with the tumor microenvironment playing a critical role in disease progression and patient outcomes. METHODS This study evaluated tumor samples from 40 female patients with luminal A and B breast cancer, utilizing flow cytometry to phenotypically characterize the immune cells and tumor cells present within the tumor tissue. RESULTS The luminal B-like tumor samples exhibited increased infiltration of CD4+ cells, regulatory T cells (Tregs), and Th17 cells and decreased levels of NK cells, γδ T cells, Th1 cells, and follicular T cells, which is indicative of a more immunosuppressive tumor microenvironment. CONCLUSIONS These findings suggest that luminal B-like tumors have a microenvironment that is less supportive of effective anti-tumor immune responses compared to luminal A tumors. This study enhances the understanding of the immunological differences between luminal subtypes of breast cancer and identifies potential new therapeutic targets and biomarkers that could drive advancements in precision medicine for breast cancer management.
Collapse
Affiliation(s)
- Tânia Moura
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Avenida Bissaya Barreto, Bloco Hospitalar de Celas, nº 205, 3000-076 Coimbra, Portugal; (T.M.); (I.S.); (S.S.)
- Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal;
| | - Olga Caramelo
- Gynecology Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal;
| | - Isabel Silva
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Avenida Bissaya Barreto, Bloco Hospitalar de Celas, nº 205, 3000-076 Coimbra, Portugal; (T.M.); (I.S.); (S.S.)
| | - Sandra Silva
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Avenida Bissaya Barreto, Bloco Hospitalar de Celas, nº 205, 3000-076 Coimbra, Portugal; (T.M.); (I.S.); (S.S.)
| | - Manuela Gonçalo
- Medical Imaging Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal; (M.G.); (M.A.P.)
| | - Maria Antónia Portilha
- Medical Imaging Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal; (M.G.); (M.A.P.)
| | - João N. Moreira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Ana M. Gil
- Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal;
- CICECO─Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Paula Laranjeira
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Avenida Bissaya Barreto, Bloco Hospitalar de Celas, nº 205, 3000-076 Coimbra, Portugal; (T.M.); (I.S.); (S.S.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Polo das Ciências da Saúde, Sub-Unidade 1, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Avenida Bissaya Barreto, Bloco Hospitalar de Celas, nº 205, 3000-076 Coimbra, Portugal; (T.M.); (I.S.); (S.S.)
- Gynecology Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal;
- Medical Imaging Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal; (M.G.); (M.A.P.)
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Instituto Politécnico de Coimbra, ESTESC—Coimbra Health School, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal
| |
Collapse
|
5
|
Zhang W, Xiao Y, Zhou Q, Zhu X, Zhang Y, Xiang Q, Wu S, Song X, Zhao J, Yuan R, Xiao B, Li L. KNSTRN Is a Prognostic Biomarker That Is Correlated with Immune Infiltration in Breast Cancer and Promotes Cell Cycle and Proliferation. Biochem Genet 2024; 62:3709-3739. [PMID: 38198023 PMCID: PMC11427568 DOI: 10.1007/s10528-023-10615-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/29/2023] [Indexed: 01/11/2024]
Abstract
Kinetochore-localized astrin/SPAG5-binding protein (KNSTRN) promotes the progression of bladder cancer and lung adenocarcinoma. However, its expression and biological function in breast cancer remain largely unknown. Therefore, this study aimed to analyze KNSTRN expression, prognoses, correlation with immune infiltration, expression-associated genes, and regulated signaling pathways to characterize its role in regulating the cell cycle using both bioinformatics and in vitro functional experiments. Analyses of The Cancer Genome Atlas, Gene Expression Omnibus, TIMER, and The Human Protein Atlas databases revealed a significant upregulation of KNSTRN transcript and protein levels in breast cancer. Kaplan-Meier survival analyses demonstrated a significant association between high expression of KNSTRN and poor overall survival, relapse-free survival, post-progression survival, and distant metastases-free survival in patients with breast cancer. Furthermore, multivariate Cox regression analyses confirmed that KNSTRN is an independent prognostic factor for breast cancer. Immune infiltration analysis indicated a positive correlation between KNSTRN expression and T regulatory cell infiltration while showing a negative correlation with Tgd and natural killer cell infiltration. Gene set enrichment analysis along with single-cell transcriptome data analysis suggested that KNSTRN promoted cell cycle progression by regulating the expression of key cell cycle proteins. The overexpression and silencing of KNSTRN in vitro, respectively, promoted and inhibited the proliferation of breast cancer cells. The overexpression of KNSTRN enhanced the expression of key cell cycle regulators, including CDK4, CDK6, and cyclin D3, thereby accelerating the G1/S phase transition and leading to aberrant proliferation of breast cancer cells. In conclusion, our study demonstrates that KNSTRN functions as an oncogene in breast cancer by regulating immune response, promoting G1/S transition, and facilitating breast cancer cell proliferation. Moreover, KNSTRN has potential as a molecular biomarker for diagnostic and prognostic prediction in breast cancer.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
- Department of Laboratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215008, China
| | - Yuhan Xiao
- School of Public Health, Dali University, Dali, 671000, China
| | - Quan Zhou
- Department of Laboratory Medicine, General Hospital of Southern Theater Command of People's Liberation Army (PLA), Guangzhou, 510010, China
| | - Xin Zhu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yanxia Zhang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Qin Xiang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Shunhong Wu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Xiaoyu Song
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Junxiu Zhao
- School of Public Health, Dali University, Dali, 671000, China
| | - Ruanfei Yuan
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Bin Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
- Department of Laboratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215008, China.
| |
Collapse
|
6
|
Camargo CP, Alapan Y, Muhuri AK, Lucas SN, Thomas SN. Single-cell adhesive profiling in an optofluidic device elucidates CD8 + T lymphocyte phenotypes in inflamed vasculature-like microenvironments. CELL REPORTS METHODS 2024; 4:100743. [PMID: 38554703 PMCID: PMC11046032 DOI: 10.1016/j.crmeth.2024.100743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 12/28/2023] [Accepted: 03/08/2024] [Indexed: 04/02/2024]
Abstract
Tissue infiltration by circulating leukocytes occurs via adhesive interactions with the local vasculature, but how the adhesive quality of circulating cells guides the homing of specific phenotypes to different vascular microenvironments remains undefined. We developed an optofluidic system enabling fluorescent labeling of photoactivatable cells based on their adhesive rolling velocity in an inflamed vasculature-mimicking microfluidic device under physiological fluid flow. In so doing, single-cell level multidimensional profiling of cellular characteristics could be characterized and related to the associated adhesive phenotype. When applied to CD8+ T cells, ligand/receptor expression profiles and subtypes associated with adhesion were revealed, providing insight into inflamed tissue infiltration capabilities of specific CD8+ T lymphocyte subsets and how local vascular microenvironmental features may regulate the quality of cellular infiltration. This methodology facilitates rapid screening of cell populations for enhanced homing capabilities under defined biochemical and biophysical microenvironments, relevant to leukocyte homing modulation in multiple pathologies.
Collapse
Affiliation(s)
- Camila P Camargo
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332, GA, USA
| | - Yunus Alapan
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332, GA, USA
| | - Abir K Muhuri
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332, GA, USA
| | - Samuel N Lucas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta 30332, GA, USA
| | - Susan N Thomas
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta 30332, GA, USA; Winship Cancer Institute, Emory University, Atlanta 30322, GA, USA.
| |
Collapse
|
7
|
Akinsipe T, Mohamedelhassan R, Akinpelu A, Pondugula SR, Mistriotis P, Avila LA, Suryawanshi A. Cellular interactions in tumor microenvironment during breast cancer progression: new frontiers and implications for novel therapeutics. Front Immunol 2024; 15:1302587. [PMID: 38533507 PMCID: PMC10963559 DOI: 10.3389/fimmu.2024.1302587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 02/16/2024] [Indexed: 03/28/2024] Open
Abstract
The breast cancer tumor microenvironment (TME) is dynamic, with various immune and non-immune cells interacting to regulate tumor progression and anti-tumor immunity. It is now evident that the cells within the TME significantly contribute to breast cancer progression and resistance to various conventional and newly developed anti-tumor therapies. Both immune and non-immune cells in the TME play critical roles in tumor onset, uncontrolled proliferation, metastasis, immune evasion, and resistance to anti-tumor therapies. Consequently, molecular and cellular components of breast TME have emerged as promising therapeutic targets for developing novel treatments. The breast TME primarily comprises cancer cells, stromal cells, vasculature, and infiltrating immune cells. Currently, numerous clinical trials targeting specific TME components of breast cancer are underway. However, the complexity of the TME and its impact on the evasion of anti-tumor immunity necessitate further research to develop novel and improved breast cancer therapies. The multifaceted nature of breast TME cells arises from their phenotypic and functional plasticity, which endows them with both pro and anti-tumor roles during tumor progression. In this review, we discuss current understanding and recent advances in the pro and anti-tumoral functions of TME cells and their implications for developing safe and effective therapies to control breast cancer progress.
Collapse
Affiliation(s)
- Tosin Akinsipe
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Rania Mohamedelhassan
- Department of Chemical Engineering, College of Engineering, Auburn University, Auburn, AL, United States
| | - Ayuba Akinpelu
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Satyanarayana R. Pondugula
- Department of Chemical Engineering, College of Engineering, Auburn University, Auburn, AL, United States
| | - Panagiotis Mistriotis
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - L. Adriana Avila
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
8
|
Camargo CP, Muhuri AK, Alapan Y, Sestito LF, Khosla M, Manspeaker MP, Smith AS, Paulos CM, Thomas SN. A dhesion analysis via a tumor vasculature-like microfluidic device identifies CD8 + T cells with enhanced tumor homing to improve cell therapy. Cell Rep 2023; 42:112175. [PMID: 36848287 DOI: 10.1016/j.celrep.2023.112175] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 12/14/2022] [Accepted: 02/13/2023] [Indexed: 02/27/2023] Open
Abstract
CD8+ T cell recruitment to the tumor microenvironment is critical for the success of adoptive cell therapy (ACT). Unfortunately, only a small fraction of transferred cells home to solid tumors. Adhesive ligand-receptor interactions have been implicated in CD8+ T cell homing; however, there is a lack of understanding of how CD8+ T cells interact with tumor vasculature-expressed adhesive ligands under the influence of hemodynamic flow. Here, the capacity of CD8+ T cells to home to melanomas is modeled ex vivo using an engineered microfluidic device that recapitulates the hemodynamic microenvironment of the tumor vasculature. Adoptively transferred CD8+ T cells with enhanced adhesion in flow in vitro and tumor homing in vivo improve tumor control by ACT in combination with immune checkpoint blockade. These results show that engineered microfluidic devices can model the microenvironment of the tumor vasculature to identify subsets of T cells with enhanced tumor infiltrating capabilities, a key limitation in ACT.
Collapse
Affiliation(s)
- Camila P Camargo
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Abir K Muhuri
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Yunus Alapan
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Lauren F Sestito
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Megha Khosla
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Margaret P Manspeaker
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Aubrey S Smith
- Winship Cancer Institute, Emory University, Atlanta, GA 30332, USA; Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - Susan N Thomas
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30332, USA.
| |
Collapse
|
9
|
Chen PC, Kaur K, Ko MW, Huerta-Yepez S, Jain Y, Jewett A. Regulation of Cytotoxic Immune Effector Function by AJ3 Probiotic Bacteria in Amyotrophic Lateral Sclerosis (ALS). Crit Rev Immunol 2023; 43:13-26. [PMID: 37522558 DOI: 10.1615/critrevimmunol.2023047231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Our recent studies indicated that amyotrophic lateral sclerosis (ALS) patients suffer from significantly elevated levels of interferon-gamma (IFN-γ) secretion by natural killer (NK) and CD8+ T cells, which may be responsible for the immune-pathologies seen in central nervous system and in peripheral organs of the patients. In order to counter such elevated induction of IFN-γ in patients we designed a treatment strategy to increase anti-inflammatory cytokine interleukin-10 (IL-10) by the use of probiotic strains which significantly increase the levels of IL-10. Therefore, in this paper we demonstrate disease specific functions of Al-Pro (AJ3) formulated for the adjunct treatment of auto-immune diseases including ALS, and compared the function with CA/I-Pro (AJ4) for the treatment of cancer and viral diseases, and NK-CLK (AJ2) for maintenance of immune balance and promotion of disease prevention. The three different formulations of probiotic bacteria have distinct profiles of activation of peripheral blood mononuclear cells (PBMCs), NK, and CD8+ T cells, and their induced activation is different from those mediated by either IL-2 or IL-2 + anti-CD16 monoclonal antibodies (mAbs) or IL-2 + anti-CD3/CD28 mAbs. IL-2 + anti-CD16 mAb activation of PBMCs and NK cells had the highest IFN-γ/IL-10 ratio, whereas IL-2 combination with sAJ4 had the next highest followed by IL-2 + sAJ2 and the lowest was seen with IL-2 + sAJ3. Accordingly, the highest secretion of IFN-γ was seen when the PBMCs and NK cells were treated with IL-2 + sAJ4, intermediate for IL-2 + sAJ2 and the lowest with IL-2 + sAJ3. The levels of IFN-γ induction and the ratio of IFN-γ to IL-10 induced by different probiotic bacteria formulation in the absence of IL-2 treatment remained much lower when compared to those treated in the presence of IL-2. Of note is the difference between NK cells and CD8+ T cells in which synergistic induction of IFN-y by IL-2 + sAJ4 was significantly higher in NK cells than those seen by CD8+ T cells. Based on these results, sAJ3 should be effective in alleviating auto-immunity seen in ALS since it will greatly regulate the levels and function of IFN-γ negatively, decreasing overactivation of cytotoxic immune effectors and prevention of death in motor neurons.
Collapse
Affiliation(s)
- Po-Chun Chen
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA
| | - Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA
| | - Meng-Wei Ko
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA
| | - Sara Huerta-Yepez
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, Los Angeles, CA 90095, USA
| | - Yash Jain
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, Los Angeles, CA 90095, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA; The Jonsson Comprehensive Cancer Center, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA
| |
Collapse
|
10
|
Prognostic value of CD8 + tumor-infiltrating T cells in patients with breast cancer: A systematic review and meta-analysis. Oncol Lett 2022; 25:39. [PMID: 36589661 PMCID: PMC9773320 DOI: 10.3892/ol.2022.13625] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/19/2022] [Indexed: 12/13/2022] Open
Abstract
CD8+ tumor-infiltrating lymphocytes have been regarded as potential biomarkers for cancer prognosis, while the prognostic effect of CD8+ tumor-infiltrating T cells remains controversial in breast cancer. In the present study, a meta-analysis was performed to evaluate the prognostic value of CD8+ T cells in breast cancer and the associations between CD8+ T cells and the pathological characteristics. The PubMed, Embase and the Cochrane Library were systematically searched entries added from the establishment of the database to November 2021 and prospective or retrospective studies of patients with breast cancer were included. The Newcastle-Ottawa Scale was used to assess the quality of evidence for each study. STATA 15.1 was used for the data analysis. A total of 14 studies comprising 22,222 patients were included in the final analysis and the pooled results suggested that a high CD8+ T-cell infiltration level was significantly related to better overall survival [hazard ratio (HR)=0.70, 95% confidence interval (CI): 0.60-0.82, P<0.001] and disease-free survival (HR=0.63, 95% CI: 0.49-0.81, P<0.001) for patients with breast cancer. In addition, a high CD8+ T-cell infiltration level was significantly associated with decreased expression of estrogen receptor [odds ratio (OR)=1.92, 95% CI: 1.30-2.85, P=0.001] and progesterone receptor (OR=1.66, 95% CI: 1.14-2.42, P=0.008), and increased human epidermal growth factor receptor 2 expression (OR=0.79, 95% CI: 0.66-0.94, P=0.010) in patients with breast cancer, while there was no significant association between CD8+ T-cell infiltration and age, tumor size or lymph node status of patients with breast cancer (P>0.05). In conclusion, CD8+ T-cell infiltration is of prognostic value in patients with breast cancer. High levels of CD8+ T-cell infiltration were related to improved prognosis, including OS and DFS, in patients with breast cancer.
Collapse
|
11
|
Li B. Why do tumor-infiltrating lymphocytes have variable efficacy in the treatment of solid tumors? Front Immunol 2022; 13:973881. [PMID: 36341370 PMCID: PMC9635507 DOI: 10.3389/fimmu.2022.973881] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/14/2022] [Indexed: 07/30/2023] Open
Abstract
Lymphocytes in tumor tissue are called tumor-infiltrating lymphocytes (TILs), and they play a key role in the control and treatment of tumor diseases. Since the discovery in 1987 that cultured TILs can kill tumor cells more than 100 times more effectively than T-cells cultured from peripheral blood in melanoma, it has been confirmed that cultured TILs can successfully cure clinical patients with melanoma. Since 1989, after we investigated TIL isolation performance from solid tumors, we modified some procedures to increase efficacy, and thus successfully established new TIL isolation and culture methods in 1994. Moreover, our laboratory and clinicians using our cultured TILs have published more than 30 papers. To improve the efficacy of TILs, we have been carrying out studies of TIL efficacy to treat solid tumor diseases for approximately 30 years. The three main questions of TIL study have been "How do TILs remain silent in solid tumor tissue?", "How do TILs attack homologous and heterologous antigens from tumor cells of solid tumors?", and "How do TILs infiltrate solid tumor tissue from a distance into tumor sites to kill tumor cells?". Research on these three issues has increasingly answered these questions. In this review I summarize the main issues surrounding TILs in treating solid tumors. This review aims to study the killing function of TILs from solid tumor tissues, thereby ultimately introducing the optimal strategy for patients suffering from solid tumors through personalized immunotherapy in the near future.
Collapse
Affiliation(s)
- Biaoru Li
- Georgia Cancer Center and Department of Pediatrics, Medical College at Georgia (GA), Augusta, GA, United States
| |
Collapse
|
12
|
Choi JM, Lim SH, Liu ZP, Lee TK, Rhee JH, Yoon MS, Min JJ, Jung S. Flagellin synergistically enhances anti-tumor effect of EGFRvIII peptide in a glioblastoma-bearing mouse brain tumor model. BMC Cancer 2022; 22:986. [PMID: 36109710 PMCID: PMC9479269 DOI: 10.1186/s12885-022-10023-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/25/2022] [Indexed: 11/25/2022] Open
Abstract
Background Glioblastoma (GBM) is the most aggressive type of brain tumor with heterogeneity and strong invasive ability. Treatment of GBM has not improved significantly despite the progress of immunotherapy and classical therapy. Epidermal growth factor receptor variant III (EGFRvIII), one of GBM-associated mutants, is regarded as an ideal therapeutic target in EGFRvIII-expressed GBM patients because it is a tumor-specific receptor expressed only in tumors. Flagellin B (FlaB) originated from Vibrio vulnificus, is known as a strong adjuvant that enhances innate and adaptive immunity in various vaccine models. This study investigated whether FlaB synergistically could enhance the anti-tumor effect of EGFRvIII peptide (PEGFRvIII). Methods EGFRvIII-GL261/Fluc cells were used for glioblastoma-bearing mouse brain model. Cell-bearing mice were inoculated with PBS, FlaB alone, PEGFRvIII alone, and PEGFRvIII plus FlaB. Tumor growth based on MRI and the survival rate was investigated. T cell population was examined by flow cytometry analysis. Both cleaved caspase-3 and CD8 + lymphocytes were shown by immunohistochemistry (IHC) staining. Results The PEGFRvIII plus FlaB group showed delayed tumor growth and increased survival rate when compared to other treatment groups. As evidence of apoptosis, cleaved caspase-3 expression and DNA disruption were more increased in the PEGFRvIII plus FlaB group than in other groups. In addition, the PEGFRvIII plus FlaB group showed more increased CD8 + T cells and decreased Treg cells than other treatment groups in the brain. Conclusions FlaB can enhance the anti-tumor effect of PEGFRvIII by increasing CD8 + T cell response in a mouse brain GBM model. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10023-6.
Collapse
|
13
|
Karimi-Shahri M, Khorramdel M, Zarei S, Attarian F, Hashemian P, Javid H. Glioblastoma, an opportunity T cell trafficking could bring for the treatment. Mol Biol Rep 2022; 49:9863-9875. [DOI: 10.1007/s11033-022-07510-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/22/2022] [Indexed: 01/22/2023]
|
14
|
Immunological profiles of the breast cancer microenvironment represented by tumor-infiltrating lymphocytes and PD-L1 expression. Sci Rep 2022; 12:8098. [PMID: 35577913 PMCID: PMC9110375 DOI: 10.1038/s41598-022-11578-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/28/2022] [Indexed: 11/29/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) and programmed cell death 1 ligand 1 (PD-L1) are established prognostic and predictive biomarkers for certain breast cancer subsets. However, their association with the immune response complexity is not fully understood. Therefore, we analyzed the association between the immune cell fractions in breast cancer tissues and histologically assessed TIL (hTIL) and PD-L1 (hPD-L1). Forty-five tumor and eighteen blood samples were collected from patients with breast cancer. Total leukocyte counts, frequency of 11 immune cell populations, and PD-L1 expression in each cell fraction were evaluated by flow cytometry. TILs and PD-L1 were assessed by hematoxylin and eosin staining and immunohistochemistry, respectively. A higher hTIL score showed association with increased leukocyte infiltration, higher CD4+ and CD8+ T cell proportions, and lower natural killer and natural killer T cell proportions. PD-L1 was highly expressed in nonclassical monocytes, monocyte/macrophages, myeloid-derived suppressor cells, myeloid dendritic cells, dendritic cells, and other lineages in tumors. hPD-L1 positivity reflected PD-L1 expression accurately in these fractions, as well as increased leukocyte infiltration in tumors. These results indicate that hTILs reflect differences in the immune responses in the tumor microenvironment, and certain immune cell fractions are favorably expressed in the PD-L1 pathway in breast cancer microenvironments.
Collapse
|
15
|
Mi J, Ye Q, Min Y. Advances in Nanotechnology Development to Overcome Current Roadblocks in CAR-T Therapy for Solid Tumors. Front Immunol 2022; 13:849759. [PMID: 35401561 PMCID: PMC8983935 DOI: 10.3389/fimmu.2022.849759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy for the treatment of hematologic tumors has achieved remarkable success, with five CAR-T therapies approved by the United States Food and Drug Administration. However, the efficacy of CAR-T therapy against solid tumors is not satisfactory. There are three existing hurdles in CAR-T cells for solid tumors. First, the lack of a universal CAR to recognize antigens at the site of solid tumors and the compact tumor structure make it difficult for CAR-T cells to locate in solid tumors. Second, soluble inhibitors and suppressive immune cells in the tumor microenvironment can inhibit or even inactivate T cells. Third, low survival and proliferation rates of CAR-T cells in vivo significantly influence the therapeutic effect. As an emerging method, nanotechnology has a great potential to enhance cell proliferation, activate T cells, and restarting the immune response. In this review, we discuss how nanotechnology can modify CAR-T cells through variable methods to improve the therapeutic effect of solid tumors.
Collapse
Affiliation(s)
- Juan Mi
- Department of Pathology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qing Ye
- Department of Pathology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuanzeng Min
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China.,Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Science at the Microscale, University of Science and Technology of China, Hefei, China
| |
Collapse
|
16
|
Simsek H, Klotzsch E. The solid tumor microenvironment-Breaking the barrier for T cells: How the solid tumor microenvironment influences T cells: How the solid tumor microenvironment influences T cells. Bioessays 2022; 44:e2100285. [PMID: 35393714 DOI: 10.1002/bies.202100285] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/20/2022]
Abstract
The tumor microenvironment (TME) plays a pivotal role in the behavior and development of solid tumors as well as shaping the immune response against them. As the tumor cells proliferate, the space they occupy and their physical interactions with the surrounding tissue increases. The growing tumor tissue becomes a complex dynamic structure, containing connective tissue, vascular structures, and extracellular matrix (ECM) that facilitates stimulation, oxygenation, and nutrition, necessary for its fast growth. Mechanical cues such as stiffness, solid stress, interstitial fluid pressure (IFP), matrix density, and microarchitecture influence cellular functions and ultimately tumor progression and metastasis. In this fight, our body is equipped with T cells as its spearhead against tumors. However, the altered biochemical and mechanical environment of the tumor niche affects T cell efficacy and leads to their exhaustion. Understanding the mechanobiological properties of the TME and their effects on T cells is key for developing novel adoptive tumor immunotherapies.
Collapse
Affiliation(s)
- Hasan Simsek
- Institute for Biology, Experimental Biophysics/Mechanobiology, Humboldt University of Berlin, Berlin, Germany
| | - Enrico Klotzsch
- Institute for Biology, Experimental Biophysics/Mechanobiology, Humboldt University of Berlin, Berlin, Germany.,Laboratory of Applied Mechanobiology, Department for Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
17
|
Zhong Y, Zhuang Z, Mo P, Lin M, Gong J, Huang J, Mo H, Lu Y, Huang M. Overexpression of SKA3 correlates with poor prognosis in female early breast cancer. PeerJ 2022; 9:e12506. [PMID: 34993016 PMCID: PMC8675262 DOI: 10.7717/peerj.12506] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 10/26/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Spindle and kinetochore associated complex subunit 3 (SKA3) plays an important role in tumorigenesis and the progression of various tumors. But the relationship between SKA3 and early breast cancer remains unclear. The study aimed to explore the prognostic significance of SKA3 in breast cancer. METHODS In the study, SKA3 expression was initially assessed using the Oncomine database and The Cancer Genome Atlas database (TCGA). Then, we presented validation results for RT-qPCR (quantitative reverse transcription PCR) and ELISA (enzyme-linked immunosorbent assay). The relationship between clinical characteristics and SKA3 expression was assessed by Chi-square test and Fisher's exact test. Kaplan-Meier method and Cox regression analysis were conducted to evaluate the prognostic value of SKA3. Gene set enrichment analysis (GSEA) was performed to screen biological pathways using the TCGA dataset. Besides, single sample gene set enrichment analysis (ssGSEA) was utilized to identify immune infiltration cells about SKA3. RESULTS SKA3 mRNA was expressed at high levels in breast cancer tissues compared with normal tissues. Chi-square test and Fisher's exact test showed SKA3 expression was related to age, tumor (T) classification, node (N) classification, tumor-node-metastasis (TNM) stage, estrogen receptor (ER), progesterone receptor (PR), molecular subtype, and race. RT-qPCR results showed that SKA3 expression was overexpressed in ER, PR status, and molecular subtype in Chinese people. Kaplan-Meier curves implicated that high SKA3 expression was related to a poor prognosis in female early breast cancer patients. Cox regression models showed that high SKA3 expression could be used as an independent risk factor for female early breast cancer. Four signaling pathways were enriched in the high SKA3 expression group, including mTORC1 signaling pathway, MYC targets v1, mitotic spindle, estrogen response early. Besides, the SKA3 expression level was associate with infiltrating levels of activated CD4 T cells and eosinophils in breast cancer. CONCLUSION High SKA3 expression correlates with poor prognosis and immune infiltrates in breast cancer. SKA3 may become a biomarker for the prognosis of breast cancer.
Collapse
Affiliation(s)
- Yue Zhong
- Guangzhou University of Chinese Medicine, Guangzhou, China.,College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenjie Zhuang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peiju Mo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mandi Lin
- Galactophore Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaqian Gong
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiarong Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haiyan Mo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuyun Lu
- Galactophore Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mei Huang
- Galactophore Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
18
|
Malla RR, Vasudevaraju P, Vempati RK, Rakshmitha M, Merchant N, Nagaraju GP. Regulatory T cells: Their role in triple-negative breast cancer progression and metastasis. Cancer 2022; 128:1171-1183. [PMID: 34990009 DOI: 10.1002/cncr.34084] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/29/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and immunogenic subtype of breast cancer. This tumorigenicity is independent of hormonal or HER2 pathways because of a lack of respective receptor expression. TNBC is extremely prone to drug resistance and early recurrence because of T-regulatory cell (Treg) infiltration into the tumor microenvironment (TME) in addition to other mechanisms like genomic instability. Tumor-infiltrating Tregs interact with both tumor and stromal cells as well as extracellular matrix components in the TME and induce an immune-suppressive phenotype. Hence, treatment of TNBC with conventional therapies remains challenging. Understanding the protective mechanism of Tregs in shielding TNBC from antitumor immune responses in the TME will pave the way for developing novel, immune-based therapeutics. The current review focuses on the role of tumor-infiltrating Tregs in tumor progression and metabolic reprogramming of the TME. The authors have extended their focus to oncotargeting Treg-mediated immune suppression in breast cancer. Because of its potential role in the TME, modulating Treg activity may provide a novel strategic intervention to combat TNBC. Both under laboratory conditions and in clinical trials, currently available anticancer drugs and natural therapeutics as potential agents for targeting Tregs are explored.
Collapse
Affiliation(s)
- Rama Rao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, India.,Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, India
| | - Padmaraju Vasudevaraju
- Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, India
| | - Rahul Kumar Vempati
- Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, India
| | - Marni Rakshmitha
- Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, India
| | - Neha Merchant
- Department of Bioscience and Biotechnology, Banasthali University, Jaipur, India
| | | |
Collapse
|
19
|
Kos K, de Visser KE. The Multifaceted Role of Regulatory T Cells in Breast Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2021; 5:291-310. [PMID: 34632244 PMCID: PMC7611782 DOI: 10.1146/annurev-cancerbio-042920-104912] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The microenvironment of breast cancer hosts a dynamic cross talk between diverse players of the immune system. While cytotoxic immune cells are equipped to control tumor growth and metastasis, tumor-corrupted immunosuppressive immune cells strive to impair effective immunity and promote tumor progression. Of these, regulatory T cells (Tregs), the gatekeepers of immune homeostasis, emerge as multifaceted players involved in breast cancer. Intriguingly, clinical observations suggest that blood and intratumoral Tregs can have strong prognostic value, dictated by breast cancer subtype. Accordingly, emerging preclinical evidence shows that Tregs occupy a central role in breast cancer initiation and progression and provide critical support to metastasis formation. Here, Tregs are not only important for immune escape but also promote tumor progression independent of their immune regulatory capacity. Combining insights into Treg biology with advances made across the rapidly growing field of immuno-oncology is expected to set the stage for the design of more effective immunotherapy strategies.
Collapse
Affiliation(s)
- Kevin Kos
- Division of Tumor Biology and Immunology, Oncode Institute, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Karin E de Visser
- Division of Tumor Biology and Immunology, Oncode Institute, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.,Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
20
|
Chen Y, Klingen TA, Aas H, Wik E, Akslen LA. Tumor-associated lymphocytes and macrophages are related to stromal elastosis and vascular invasion in breast cancer. J Pathol Clin Res 2021; 7:517-527. [PMID: 34076969 PMCID: PMC8363927 DOI: 10.1002/cjp2.226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/25/2021] [Accepted: 05/05/2021] [Indexed: 02/04/2023]
Abstract
The tumor microenvironment plays a critical role in breast cancer progression. Here, we investigated tumor-infiltrating lymphocytes (TILs) and associations with macrophage numbers, tumor stromal elastosis, vascular invasion, and tumor detection mode. We performed a population-based retrospective study using data from The Norwegian Breast Cancer Screening Program in Vestfold County (2004-2009), including 200 screen-detected and 82 interval cancers. The number of TILs (CD45+, CD3+, CD4+, CD8+, and FOXP3+) and tumor-associated macrophages (CD163+) was counted using immunohistochemistry on tissue microarray slides. Lymphatic and blood vessel invasion (LVI and BVI) were recorded using D2-40 and CD31 staining, and the amount of elastosis (high/low) was determined on regular HE-stained slides. High numbers of all TIL subsets were associated with LVI (p ≤ 0.04 for all), and high counts of several TIL subgroups (CD8+, CD45+, and FOXP3+) were associated with BVI (p ≤ 0.04 for all). Increased levels of all TIL subsets, except CD4+, were associated with estrogen receptor-negative tumors (p < 0.001) and high tumor cell proliferation by Ki67 (p < 0.001). Furthermore, high levels of all TIL subsets were associated with high macrophage counts (p < 0.001) and low-grade stromal elastosis (p ≤ 0.02). High counts of CD3+, CD8+, and FOXP3+ TILs were associated with interval detected tumors (p ≤ 0.04 for all). Finally, in the luminal A subgroup, high levels of CD3+ and FOXP3+ TILs were associated with shorter recurrence-free survival, and high counts of FOXP3+ were linked to reduced breast cancer-specific survival. In conclusion, higher levels of different TIL subsets were associated with stromal features such as high macrophage counts (CD163+), presence of vascular invasion, absence of stromal elastosis, as well as increased tumor cell proliferation and interval detection mode. Our findings support a link between immune cells and vascular invasion in more aggressive breast cancer. Notably, presence of TIL subsets showed prognostic value within the luminal A category.
Collapse
Affiliation(s)
- Ying Chen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of PathologyVestfold HospitalTønsbergNorway
- Department of PathologyOslo University HospitalOsloNorway
| | - Tor Audun Klingen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of PathologyVestfold HospitalTønsbergNorway
| | - Hans Aas
- Department of SurgeryVestfold HospitalTønsbergNorway
| | - Elisabeth Wik
- Centre for Cancer Biomarkers CCBIO, Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of PathologyHaukeland University HospitalBergenNorway
| | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of PathologyHaukeland University HospitalBergenNorway
| |
Collapse
|
21
|
Tang WJ, Kong QC, Cheng ZX, Liang YS, Jin Z, Chen LX, Hu WK, Liang YY, Wei XH, Guo Y, Jiang XQ. Performance of radiomics models for tumour-infiltrating lymphocyte (TIL) prediction in breast cancer: the role of the dynamic contrast-enhanced (DCE) MRI phase. Eur Radiol 2021; 32:864-875. [PMID: 34430998 DOI: 10.1007/s00330-021-08173-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/20/2021] [Accepted: 06/25/2021] [Indexed: 01/26/2023]
Abstract
OBJECTIVE To systematically investigate the effect of imaging features at different DCE-MRI phases to optimise a radiomics model based on DCE-MRI for the prediction of tumour-infiltrating lymphocyte (TIL) levels in breast cancer. MATERIALS AND METHODS This study retrospectively collected 133 patients with pathologically proven breast cancer, including 73 patients with low TIL levels and 60 patients with high TIL levels. The volumes of breast cancer lesions were manually delineated on T2-weighted imaging (T2WI), diffusion-weighted imaging (DWI), and each phase of DCE-MRI, followed by 6250 quantitative feature extractions. The least absolute shrinkage and selection operator (LASSO) method was used to select predictive feature sets for the classifiers. Four models were developed for predicting TILs: (1) single enhanced phase radiomics models; (2) fusion enhanced multi-phase radiomics models; (3) fusion multi-sequence radiomics models; and (4) a combined radiomics-based clinical model. RESULTS Image features extracted from the delayed phase MRI, especially DCE_Phase 6 (DCE_P6), demonstrated dominant predictive performances over features from other phases. The fusion multi-sequence radiomics model and combined radiomics-based clinical model achieved the highest predictive performances with areas under the curve (AUCs) of 0.934 and 0.950, respectively; however, the differences were not statistically significant. CONCLUSION The DCE-MRI radiomics model, especially image features extracted from the delayed phases, can help improve the performance in predicting TILs. The radiomics nomogram is effective in predicting TILs in breast cancer. KEY POINTS • Radiomics features extracted from DCE-MRI, especially delayed phase images, help predict TIL levels in breast cancer. • We developed a nomogram based on MRI to predict TILs in breast cancer that achieved the highest AUC of 0.950.
Collapse
Affiliation(s)
- Wen-Jie Tang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Qing-Cong Kong
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China
| | - Zi-Xuan Cheng
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Yun-Shi Liang
- Department of Pathology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Zhe Jin
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Lei-Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Wen-Ke Hu
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Ying-Ying Liang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Xin-Hua Wei
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Yuan Guo
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China.
| | - Xin-Qing Jiang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China.
| |
Collapse
|
22
|
Boosz P, Pfister F, Stein R, Friedrich B, Fester L, Band J, Mühlberger M, Schreiber E, Lyer S, Dudziak D, Alexiou C, Janko C. Citrate-Coated Superparamagnetic Iron Oxide Nanoparticles Enable a Stable Non-Spilling Loading of T Cells and Their Magnetic Accumulation. Cancers (Basel) 2021; 13:4143. [PMID: 34439296 PMCID: PMC8394404 DOI: 10.3390/cancers13164143] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 02/07/2023] Open
Abstract
T cell infiltration into a tumor is associated with a good clinical prognosis of the patient and adoptive T cell therapy can increase anti-tumor immune responses. However, immune cells are often excluded from tumor infiltration and can lack activation due to the immune-suppressive tumor microenvironment. To make T cells controllable by external forces, we loaded primary human CD3+ T cells with citrate-coated superparamagnetic iron oxide nanoparticles (SPIONs). Since the efficacy of magnetic targeting depends on the amount of SPION loading, we investigated how experimental conditions influence nanoparticle uptake and viability of cells. We found that loading in the presence of serum improved both the colloidal stability of SPIONs and viability of T cells, whereas stimulation with CD3/CD28/CD2 and IL-2 did not influence nanoparticle uptake. Furthermore, SPION loading did not impair cytokine secretion after polyclonal stimulation. We finally achieved 1.4 pg iron loading per cell, which was both located intracellularly in vesicles and bound to the plasma membrane. Importantly, nanoparticles did not spill over to non-loaded cells. Since SPION-loading enabled efficient magnetic accumulation of T cells in vitro under dynamic conditions, we conclude that this might be a good starting point for the investigation of in vivo delivery of immune cells.
Collapse
Affiliation(s)
- Philipp Boosz
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
- Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Felix Pfister
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Rene Stein
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Bernhard Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Lars Fester
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
| | - Julia Band
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Marina Mühlberger
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Eveline Schreiber
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany;
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Medical Immunology Campus Erlangen, 91054 Erlangen, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| |
Collapse
|
23
|
Zahran AM, El-Badawy O, Kamel LM, Rayan A, Rezk K, Abdel-Rahim MH. Accumulation of Regulatory T Cells in Triple Negative Breast Cancer Can Boost Immune Disruption. Cancer Manag Res 2021; 13:6019-6029. [PMID: 34377021 PMCID: PMC8349183 DOI: 10.2147/cmar.s285128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022] Open
Abstract
Background and Aim The present study was conducted to evaluate the number of Tregs in triple negative breast cancer (TNBC), in normal breast parenchyma and in the peripheral blood of these patients and controls, in addition to their correlations with the clinico-pathologic features and the outcomes of TNBC. Methods Thirty adult treatment-naïve women with non-metastatic TNBC were recruited. In addition, 20 ages matched healthy females participated as a control group. Peripheral blood samples were collected from all participants in tubes containing heparin, fresh tumor tissues were also obtained from all patients undergoing surgery, and 20 normal breast tissue samples were obtained from the same patients’ areas adjacent to the safety margins; all these samples were taken for flow cytometric detection of Tregs. Results The mean percentages of CD4+CD25+highT cells and Tregs were higher in TNBC peripheral blood than healthy controls and in malignant tissue than normal tissue. Moreover, the frequencies of tumor-infiltrating CD4+T cells and Tregs were exceeding those in the peripheral blood of cancer patients. Only tumor-infiltrating Tregs have shown increasing levels with the increase in the tumor size and were significantly higher in patients with local recurrences than those without recurrence. In addition, Tregs showed significant inverse relation with DFS and direct relation with the level of the peripheral Tregs. Conclusion The findings of the current study support the possibility that TNBC microenvironment conveys specific characteristics on Tregs distinguishing them from those in normal breast tissue or Tregs in peripheral blood, improving the capabilities of tumor-infiltrating Tregs to enhance tumor growth, local recurrence and reduce the DFS.
Collapse
Affiliation(s)
- Asmaa M Zahran
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Omnia El-Badawy
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Lamiaa M Kamel
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Amal Rayan
- Clinical Oncology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Khalid Rezk
- Surgical Oncology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Mona H Abdel-Rahim
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
24
|
Nodal status in luminal A invasive breast cancer: relationships with cytotoxic CD8 + and regulatory FOXP3 + cells tumor-associated infiltrate and other prognostic factors. Virchows Arch 2021; 479:871-882. [PMID: 34117905 PMCID: PMC8572830 DOI: 10.1007/s00428-021-03126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/24/2022]
Abstract
Luminal A breast cancers are generally associated with low metastatic potential and good prognosis. However, there is a proportion of patients, who present with metastases in lymph nodes. The aim of our study was to determine the association between the number of positive lymph nodes and infiltrates of tumor-associated cytotoxic CD8 + (CTLs), regulatory FOXP3 + T cells (Tregs), as well as other prognostic factors. Immunohistochemistry (IHC) for CD8 + and FOXP3 + was performed in 87 formalin-fixed paraffin-embedded primary breast cancer tissues, and cell infiltrate was assessed under light microscope. We observed that node-positive cases were associated with higher numbers of Treg cells and lower CTL/Treg ratio. There was also an inverse correlation between the CTL/Treg ratio and the number of metastatic lymph nodes. Similar relationships were found between the number of metastatic lymph nodes and Treg density or CTL/Treg ratio in pT1 BC. An elevated intratumoral CTL/Treg ratio was associated with pN0 stage. The relationship between lymphovascular invasion (LVI) and Treg density was also noted in node-negative tumors. In addition, more advanced nodal stage was related to LVI, higher pT, and lower PR expression. The numbers of CD8 + and FOXP3 + were also associated with tumor size, histologic grade, PR expression, and mitotic index. The results of our study suggested that the levels of tumor-infiltrating regulatory and cytotoxic cells as well as the balance between them play a role in lymphovascular spread of luminal A breast cancers.
Collapse
|
25
|
Zhang L, Li Y, Wang X, Ping Y, Wang D, Cao Y, Dai Y, Liu W, Tao Z. Five-gene signature associating with Gleason score serve as novel biomarkers for identifying early recurring events and contributing to early diagnosis for Prostate Adenocarcinoma. J Cancer 2021; 12:3626-3647. [PMID: 33995639 PMCID: PMC8120165 DOI: 10.7150/jca.52170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Compared to non-recurrent type, recurrent prostate adenocarcinoma (PCa) is highly fatal, and significantly shortens the survival time of affected patients. Early and accurate laboratory diagnosis is particularly important in identifying patients at high risk of recurrence, necessary for additional systemic intervention. We aimed to develop efficient and accurate diagnostic and prognostic biomarkers for new PCa following radical therapy. Methods: We identified differentially expressed genes (DEGs) and clinicopathological data of PCa patients from Gene Expression Omnibus (GEO) datasets and The Cancer Genome Atlas (TCGA) repositories. We then uncovered the most relevant clinical traits and genes modules associated with PCa prognosis using the Weighted gene correlation network analysis (WGCNA). Univariate Cox regression analysis and multivariate Cox proportional hazards (Cox-PH) models were performed to identify candidate gene signatures related to Disease-Free Interval (DFI). Data for internal and external cohorts were utilized to test and validate the accuracy and clinical utility of the prognostic models. Results: We constructed and validated an accurate and reliable model for predicting the prognosis of PCa using 5 Gleason score-associated gene signatures (ZNF695, CENPA, TROAP, BIRC5 and KIF20A). The ROC and Kaplan-Meier analysis revealed the model was highly accurate in diagnosing and predicting the recurrence and metastases of PCa. The accuracy of the model was validated using the calibration curves based on internal TCGA cohort and external GEO cohort. Using the model, patients could be prognostically stratified in to various groups including TNM classification and Gleason score. Multivariate analysis revealed the model could independently predict the prognosis of PCa patients and its utility was superior to that of clinicopathological characteristics. In addition, we fund the expression of the 5 gene signatures strongly and positively correlated with tumor purity but negatively correlated with infiltration CD8+ T cells to the tumor microenvironment. Conclusions: A 5 gene signatures can accurately be used in the diagnosis and prediction of PCa prognosis. Thus this can guide the treatment and management prostate adenocarcinoma.
Collapse
Affiliation(s)
- Lingyu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yu Li
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui 233030, China
| | - Xuchu Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ying Ping
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Danhua Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ying Cao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yibei Dai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| |
Collapse
|
26
|
Asiry S, Kim G, Filippou PS, Sanchez LR, Entenberg D, Marks DK, Oktay MH, Karagiannis GS. The Cancer Cell Dissemination Machinery as an Immunosuppressive Niche: A New Obstacle Towards the Era of Cancer Immunotherapy. Front Immunol 2021; 12:654877. [PMID: 33927723 PMCID: PMC8076861 DOI: 10.3389/fimmu.2021.654877] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Although cancer immunotherapy has resulted in unpreceded survival benefits to subsets of oncology patients, accumulating evidence from preclinical animal models suggests that the immunosuppressive tumor microenvironment remains a detrimental factor limiting benefit for many patient subgroups. Recent efforts on lymphocyte-mediated immunotherapies are primarily focused on eliminating cancer foci at primary and metastatic sites, but few studies have investigated the impact of these therapies on the highly complex process of cancer cell dissemination. The metastatic cascade involves the directional streaming of invasive/migratory tumor cells toward specialized blood vessel intravasation gateways, called TMEM doorways, to the peripheral circulation. Importantly, this process occurs under the auspices of a specialized tumor microenvironment, herewith referred to as "Dissemination Trajectory", which is supported by an ample array of tumor-associated macrophages (TAMs), skewed towards an M2-like polarization spectrum, and which is also vital for providing microenvironmental cues for cancer cell invasion, migration and stemness. Based on pre-existing evidence from preclinical animal models, this article outlines the hypothesis that dissemination trajectories do not only support the metastatic cascade, but also embody immunosuppressive niches, capable of providing transient and localized immunosubversion cues to the migratory/invasive cancer cell subpopulation while in the act of departing from a primary tumor. So long as these dissemination trajectories function as "immune deserts", the migratory tumor cell subpopulation remains efficient in evading immunological destruction and seeding metastatic sites, despite administration of cancer immunotherapy and/or other cytotoxic treatments. A deeper understanding of the molecular and cellular composition, as well as the signaling circuitries governing the function of these dissemination trajectories will further our overall understanding on TAM-mediated immunosuppression and will be paramount for the development of new therapeutic strategies for the advancement of optimal cancer chemotherapies, immunotherapies, and targeted therapies.
Collapse
Affiliation(s)
- Saeed Asiry
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| | - Gina Kim
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| | - Panagiota S. Filippou
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
- National Horizons Centre, Teesside University, Darlington, United Kingdom
| | - Luis Rivera Sanchez
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
| | - David Entenberg
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| | - Douglas K. Marks
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY, United States
| | - Maja H. Oktay
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| | - George S. Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| |
Collapse
|
27
|
Thike AA, Chen X, Koh VCY, Binte Md Nasir ND, Yeong JPS, Bay BH, Tan PH. Higher densities of tumour-infiltrating lymphocytes and CD4 + T cells predict recurrence and progression of ductal carcinoma in situ of the breast. Histopathology 2021; 76:852-864. [PMID: 31883279 DOI: 10.1111/his.14055] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 12/14/2019] [Accepted: 12/24/2019] [Indexed: 12/13/2022]
Abstract
AIMS Host immunity influences cancer progression and therapeutic response. We investigated the potential of tumour-infiltrating lymphocytes (TILs) around ductal carcinoma in situ (DCIS) in predicting recurrence and progression. METHODS AND RESULTS CD4, CD8, programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) expression in DCIS from 198 patients was determined by immunohistochemistry. We correlated disease-free survival (DFS), clinicopathological parameters and biomarker expression with TIL density and CD4/CD8 ratio. High TIL density was associated with high nuclear grade (P < 0.001), DCIS PD-L1 expression (P = 0.008), TIL PD-L1 expression (P < 0.001), oestrogen (ER) negativity (P < 0.001), progesterone (PR) negativity (P < 0.001), human epidermal growth factor receptor 2 (HER2) positivity (P = 0.002) and triple negativity (P = 0.001). TIL PD-L1 expression was associated with triple-negative DCIS (P = 0.028). TIL density was associated with molecular subtypes (P < 0.001). High CD4+ T cell density was associated with high nuclear grade (P = 0.001), microinvasion (P = 0.037), ER negativity (P < 0.001), PR negativity (P = 0.001), HER2 positivity (P = 0.004), triple negativity (P = 0.023) and PD-L1 expression in TILs (P < 0.011). High CD4/CD8 ratio was associated with PD-L1 expression in DCIS (P = 0.035) and TILs (P < 0.001). DCIS with higher TIL density disclosed worse DFS (P = 0.012) and was affirmed with multivariate analysis [95% confidence interval (CI) = 1.109-2.554, hazard ratio (HR) = 1.683, P = 0.014]. Poorer DFS for ipsilateral invasive recurrence was found for DCIS with higher CD4+ T cell density (P = 0.006) or CD4/CD8 ratio (P = 0.02), confirmed by multivariate analysis for the former (95% CI = 1.369-10.196, HR = 3.736, P = 0.01) and latter (95% CI = 1.311-7.935, HR = 3.225, P = 0.011). CONCLUSION DCIS with higher TIL density was associated with poorer prognostic parameters and predicted recurrence, while both CD4+ T cell density and CD4/CD8 ratio were associated with both recurrence and ipsilateral invasive recurrence.
Collapse
Affiliation(s)
- Aye Aye Thike
- Department of Anatomical Pathology, Singapore General Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | - Xiaoyang Chen
- Department of Anatomical Pathology, Singapore General Hospital, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | - Joe P S Yeong
- Department of Anatomical Pathology, Singapore General Hospital, Singapore.,Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Puay Hoon Tan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore.,Duke-NUS Medical School, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Division of Pathology, Singapore General Hospital, Singapore
| |
Collapse
|
28
|
Dai D, Liu L, Huang H, Chen S, Chen B, Cao J, Luo X, Wang F, Luo R, Liu J. Nomograms to Predict the Density of Tumor-Infiltrating Lymphocytes in Patients With High-Grade Serous Ovarian Cancer. Front Oncol 2021; 11:590414. [PMID: 33718143 PMCID: PMC7946970 DOI: 10.3389/fonc.2021.590414] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022] Open
Abstract
Background Tumor-infiltrating lymphocytes (TILs) have important roles in predicting tumor therapeutic responses and progression, however, the method of evaluating TILs is complicated. We attempted to explore the association of TILs with clinicopathological characteristics and blood indicators, and to develop nomograms to predict the density of TILs in patients with high-grade serous ovarian cancer (HGSOC). Methods The clinical profiles of 197 consecutive postoperative HGSOC patients were retrospectively analyzed. Tumor tissues and matched normal fallopian tubes were immunostained for CD3+, CD8+, and CD4+ T cells on corresponding tissue microarrays and the numbers of TILs were counted using the NIH ImageJ software. The patients were classified into low- or high-density groups for each marker (CD3, CD4, CD8). The associations of the investigated TILs to clinicopathological characteristics and blood indicators were assessed and the related predictors for densities of TILs were used to develop nomograms; which were then further evaluated using the C-index, receiver operating characteristic (ROC) curves and calibration plots. Results Menopausal status, estrogen receptor (ER), Ki-67 index, white blood cell (WBC), platelets (PLT), lactate dehydrogenase (LDH), and carbohydrate antigen 153 (CA153) had significant association with densities of tumor-infiltrating CD3+, CD8+, or CD4+ T cells. The calibration curves of the CD3+ (C-index = 0.748), CD8+ (C-index = 0.683) and CD4+ TILs nomogram (C-index = 0.759) demonstrated excellent agreement between predictions and actual observations. ROC curves of internal validation indicated good discrimination for the CD8+ TILs nomogram [area under the curve (AUC) = 0.659, 95% CI 0.582–0.736] and encouraging performance for the CD3+ (AUC= 0.708, 95% CI 0.636–0.781) and CD4+ TILs nomogram (AUC = 0.730, 95% CI 0.659–0.801). Conclusion Menopausal status, ER, Ki-67 index, WBC, PLT, LDH, and CA153 could reflect the densities of T cells in the tumor microenvironment. Novel nomograms are conducive to monitor the immune status of patients with HGSOC and help doctors to formulate the appropriate treatment strategies.
Collapse
Affiliation(s)
- Danian Dai
- Department of Gynecology and Obstetrics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.,Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lili Liu
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - He Huang
- Department of Gynecology and Obstetrics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Shangqiu Chen
- Department of Gynecology and Obstetrics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Bo Chen
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Junya Cao
- Department of Gynecology and Obstetrics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Xiaolin Luo
- Department of Gynecology and Obstetrics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Feng Wang
- Department of Gynecology and Obstetrics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Rongzhen Luo
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jihong Liu
- Department of Gynecology and Obstetrics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.,Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
29
|
Stenström J, Hedenfalk I, Hagerling C. Regulatory T lymphocyte infiltration in metastatic breast cancer-an independent prognostic factor that changes with tumor progression. Breast Cancer Res 2021; 23:27. [PMID: 33602289 PMCID: PMC7893927 DOI: 10.1186/s13058-021-01403-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/01/2021] [Indexed: 02/08/2023] Open
Abstract
Background Patients diagnosed with metastatic breast cancer have poor outcome with a median survival of approximately 2 years. While novel therapeutic options are urgently needed, the great majority of breast cancer research has focused on the primary tumor and less is known about metastatic breast cancer and the prognostic impact of the metastatic tumor microenvironment. Here we investigate the immune landscape in unique clinical material. We explore how the immune landscape changes with metastatic progression and elucidate the prognostic role of immune cells infiltrating primary tumors and corresponding lymph node and more importantly distant metastases. Methods Immunohistochemical staining was performed on human breast cancer tissue microarrays from primary tumors (n = 231), lymph node metastases (n = 129), and distant metastases (n = 43). Infiltration levels of T lymphocytes (CD3+), regulatory T lymphocytes (Tregs, FOXP3+), macrophages (CD68+), and neutrophils (NE+) were assessed in primary tumors. T lymphocytes and Tregs were further investigated in lymph node and distant metastases. Results T lymphocyte and Treg infiltration were the most clinically important immune cell populations in primary tumors. Infiltration of T lymphocytes and Tregs in primary tumors correlated with proliferation (P = 0.007, P = 0.000) and estrogen receptor negativity (P = 0.046, P = 0.026). While both T lymphocyte and Treg infiltration had a negative correlation to luminal A subtype (P = 0.031, P = 0.000), only Treg infiltration correlated to luminal B (P = 0.034) and triple-negative subtype (P = 0.019). In primary tumors, infiltration of T lymphocytes was an independent prognostic factor for recurrence-free survival (HR = 1.77, CI = 1.01–3.13, P = 0.048), while Treg infiltration was an independent prognostic factor for breast cancer-specific survival (HR = 1.72, CI = 1.14–2.59, P = 0.01). Moreover, breast cancer patients with Treg infiltration in their distant metastases had poor post-recurrence survival (P = 0.039). Treg infiltration levels changed with metastatic tumor progression in 50% of the patients, but there was no significant trend toward neither lower nor higher infiltration. Conclusion Treg infiltration could have clinical applicability as a prognostic biomarker, deciphering metastatic breast cancer patients with worse prognosis, and accordingly, could be a suitable immunotherapeutic target for patients with metastatic breast cancer. Importantly, half of the patients had changes in Treg infiltration during the course of metastatic progression emphasizing the need to characterize the metastatic immune landscape. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-021-01403-0.
Collapse
Affiliation(s)
- Jenny Stenström
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, SE-221 85, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, SE-221 85, Lund, Sweden
| | - Catharina Hagerling
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, SE-221 85, Lund, Sweden.
| |
Collapse
|
30
|
Role of CD4- and CD8-Positive T Cells in Breast Cancer Progression and Outcome: A Pilot Study of 47 Cases in Central India Region. INDIAN JOURNAL OF GYNECOLOGIC ONCOLOGY 2020. [DOI: 10.1007/s40944-020-00454-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
31
|
Wang X, Wu Z, Qiu W, Chen P, Xu X, Han W. Programming CAR T cells to enhance anti-tumor efficacy through remodeling of the immune system. Front Med 2020; 14:726-745. [PMID: 32794014 DOI: 10.1007/s11684-020-0746-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor (CAR) T cells have been indicated effective in treating B cell acute lymphoblastic leukemia and non-Hodgkin lymphoma and have shown encouraging results in preclinical and clinical studies. However, CAR T cells have achieved minimal success against solid malignancies because of the additional obstacles of their insufficient migration into tumors and poor amplification and persistence, in addition to antigen-negative relapse and an immunosuppressive microenvironment. Various preclinical studies are exploring strategies to overcome the above challenges. Mobilization of endogenous immune cells is also necessary for CAR T cells to obtain their optimal therapeutic effect given the importance of the innate immune responses in the elimination of malignant tumors. In this review, we focus on the recent advances in the engineering of CAR T cell therapies to restore the immune response in solid malignancies, especially with CAR T cells acting as cellular carriers to deliver immunomodulators to tumors to mobilize the endogenous immune response. We also explored the sensitizing effects of conventional treatment approaches, such as chemotherapy and radiotherapy, on CAR T cell therapy. Finally, we discuss the combination of CAR T cells with biomaterials or oncolytic viruses to enhance the anti-tumor outcomes of CAR T cell therapies in solid tumors.
Collapse
Affiliation(s)
- Xiaohui Wang
- College of Biotechnology, Southwest University, Chongqing, 400715, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhiqiang Wu
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei Qiu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China
| | - Ping Chen
- College of Biotechnology, Southwest University, Chongqing, 400715, China
| | - Xiang Xu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China.
| | - Weidong Han
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
32
|
Kaur K, Ko MW, Ohanian N, Cook J, Jewett A. Osteoclast-expanded super-charged NK-cells preferentially select and expand CD8+ T cells. Sci Rep 2020; 10:20363. [PMID: 33230147 PMCID: PMC7683603 DOI: 10.1038/s41598-020-76702-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023] Open
Abstract
Osteoclasts (OCs) and much less dendritic cells (DCs) induce significant expansion and functional activation of NK cells, and furthermore, the OC-expanded NK cells preferentially increase the expansion and activation of CD8+ T cells by targeting CD4+ T cells. When autologous OCs were used to expand patient NK cells much lower percentages of expanded CD8+ T cells, decreased numbers of expanded NK cells and decreased functions of NK cells could be observed, and the addition of allogeneic healthy OCs increased the patients' NK function. Mechanistically, OC-expanded NK cells were found to lyse CD4+ T cells but not CD8+ T cells suggesting potential selection of CD8+ T cells before their expansion by OC activated NK cells. In agreement, Increased IFN-γ secretion, and NK cell-mediated cytotoxicity and higher percentages of CD8+ T cells, in various tissue compartments of oral tumor-bearing hu-BLT mice in response to immunotherapy by OC-expanded NK cells were observed. Thus, our results indicate an important relationship between NK and CD8+ T cells.
Collapse
Affiliation(s)
- Kawaljit Kaur
- Division of Oral Biology and Oral Medicine, School of Dentistry and Medicine, Los Angeles, CA, USA
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Meng-Wei Ko
- Division of Oral Biology and Oral Medicine, School of Dentistry and Medicine, Los Angeles, CA, USA
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Nick Ohanian
- Division of Oral Biology and Oral Medicine, School of Dentistry and Medicine, Los Angeles, CA, USA
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Jessica Cook
- Division of Oral Biology and Oral Medicine, School of Dentistry and Medicine, Los Angeles, CA, USA
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Anahid Jewett
- Division of Oral Biology and Oral Medicine, School of Dentistry and Medicine, Los Angeles, CA, USA.
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry and Medicine, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA.
- The Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
33
|
Pötzsch M, Berg E, Hummel M, Stein U, von Winterfeld M, Jöhrens K, Rau B, Daum S, Treese C. Better prognosis of gastric cancer patients with high levels of tumor infiltrating lymphocytes is counteracted by PD-1 expression. Oncoimmunology 2020; 9:1824632. [PMID: 33101772 PMCID: PMC7553533 DOI: 10.1080/2162402x.2020.1824632] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 12/20/2022] Open
Abstract
The prognostic potential of anti-tumor immune responses is becoming increasingly important in adenocarcinoma of the gastroesophageal junction and stomach (AGE/S) especially regarding the use of immune checkpoint inhibitors. This study analyzes for the first time the prognostic impact of tumor-infiltrating lymphocytes (TILs) and checkpoint inhibitors in a large Caucasian cohort in patients with AGE/S. We screened tissue samples from 438 therapy-naïve patients with AGE/S undergoing surgery between 1992 and 2005, examined in a tissue microarray (TMA) and stained against human CD3, CD4, CD8, PD-1, and PD-L1. Out of 438 tissue samples, 210 were eligible for multivariate analysis. This revealed that high infiltration with CD3+, CD4+, or CD8+ TILs was associated with an increased overall survival in AGE/S patients, which could only be confirmed in multivariate analysis for CD3 (HR: 0.326; p = .023). Independent improved survival was limited to gastric cancer patients and to early tumor stages as long as TILs did not express PD-1 (HR: 1.522; p = .021). Subgroup analyses indicate that TIL-dependent anti-tumor immune response is only effective in gastric cancer patients in early stages of disease in PD-1 negative TILs. Combined analysis of PD-1 and CD3 could serve as a prognostic marker for the clinical outcome of gastric cancer patients and could also be of interest for immunotherapy.
Collapse
Affiliation(s)
- M. Pötzsch
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
| | - E. Berg
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
| | - M. Hummel
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- 1.Institute for Pathology, Charité - Universitätsmedizin Berlin, Charité Campus Mitte, Berlin, Germany
| | - U. Stein
- Experimental and Clinical Research Center, Charité - Universitätsmedizin, Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - M. von Winterfeld
- Institute of Pathology Heidelberg, University Hospital Heidelberg, Germany
| | - K. Jöhrens
- Institute of Pathology, University Carl Gustav Carus, Dresden, Germany
| | - B. Rau
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of Surgery, Campus Virchow-Klinikum and Charité Campus Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - S. Daum
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - C. Treese
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
- Experimental and Clinical Research Center, Charité - Universitätsmedizin, Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
34
|
Van Bockstal MR, Noel F, Guiot Y, Duhoux FP, Mazzeo F, Van Marcke C, Fellah L, Ledoux B, Berlière M, Galant C. Predictive markers for pathological complete response after neo-adjuvant chemotherapy in triple-negative breast cancer. Ann Diagn Pathol 2020; 49:151634. [PMID: 32987254 DOI: 10.1016/j.anndiagpath.2020.151634] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022]
Abstract
A combination of Sox10 and GATA3 was previously identified as a marker for metastatic triple-negative breast cancer (TNBC), but it is uncertain whether their expression is associated with pathological complete response (pCR) after neoadjuvant chemotherapy (NAC). This study investigates the predictive value of clinicopathological characteristics, as well as protein expression of Sox10, GATA3, p53 and p63, in a consecutive series of TNBC patients treated with NAC. Archived hematoxylin & eosin stained slides of core biopsies and resection specimens from 35 TNBC patients were reviewed. The following clinicopathological characteristics were determined at the biopsy level: age at diagnosis, cancer type, Nottingham grade, lympho-vascular invasion, syncytial growth, necrosis, clear cell differentiation, myxoid peritumor stroma, stromal tumor-infiltrating lymphocytes (sTILs) and presence of an in situ component. The MD Anderson residual cancer burden (RCB) score and corresponding RCB class were determined. Immunohistochemistry for Sox10, p53, GATA3 and p63 was performed at the biopsy level. sTILs, either as a continuous or as a dichotomous variable, were the only parameter that was significantly associated with pCR in univariable and multivariable analyses. Assessment of sTILs showed moderate to good interobserver agreement. High sTILs (≥40%) were significantly associated with increased pCR rates, and this association was observer-independent. This retrospective study of a consecutive community-based cohort of TNBC patients confirms that sTILs are a robust, observer-independent predictor for therapeutic response after NAC. The combination of Sox10, GATA3 and p53 immunoreactivity is unlikely to harbor any predictive value for pCR in TNBC.
Collapse
Affiliation(s)
- Mieke R Van Bockstal
- Department of Pathology, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium; Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium; Breast Clinic, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium.
| | - Fanchon Noel
- Department of Pathology, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Yves Guiot
- Department of Pathology, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Francois P Duhoux
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium; Breast Clinic, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Filomena Mazzeo
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium; Breast Clinic, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Cédric Van Marcke
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium; Breast Clinic, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Latifa Fellah
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium; Breast Clinic, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of Radiology, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Benjamin Ledoux
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium; Breast Clinic, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium; Department of Oncologic Radiotherapy, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Martine Berlière
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium; Breast Clinic, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Christine Galant
- Department of Pathology, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium; Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium; Breast Clinic, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| |
Collapse
|
35
|
Oner G, Altintas S, Canturk Z, Tjalma W, Van Berckelaer C, Broeckx G, Zwaenepoel K, Tholhuijsen M, Verhoeven Y, Berneman Z, Peeters M, Pauwels P, van Dam PA. The immunologic aspects in hormone receptor positive breast cancer. Cancer Treat Res Commun 2020; 25:100207. [PMID: 32896829 DOI: 10.1016/j.ctarc.2020.100207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/04/2020] [Accepted: 07/05/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Although hormone receptor positive/HER2-negative (HR +/HER2-) breast cancer is the most diagnosed breast cancer type, the immunologic aspects HR positive breast cancer (BC) has been neglected until recently. The purpose of this paper is to review the current knowledge of the immune environment in HR positive BC and the potential use of immunotherapy in these patients. METHOD A computer-based literature research was carried out using PubMed, American Society of Clinical Oncology Annual Meeting (ASCO) and San Antonio Breast Cancer Symposium (SABCS). RESULTS The tumour microenvironment (TME), with infiltrating immune cells, plays an important role in HR positive BC. However, the effects of these immune cells are different in the luminal cancers compared to the other breast cancer types. Even though PD-1 and PD-L1 are less expressed in HR positive BC, pathological complete response (pCR) was more often seen after PD-1 inhibitor treatment in patients with an increased expression. The studies support the assertion that endocrine therapy has immunomodulatory effect. CONCLUSION The reviewed literature indicates that immune cells play an important role in HR positive BC. Considerably more research is needed to determine the real effect of the TME in this patient group.
Collapse
Affiliation(s)
- Gizem Oner
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium; Department of General Surgery, Kocaeli University, Kocaeli, Turkey.
| | - Sevilay Altintas
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Zafer Canturk
- Department of General Surgery, Kocaeli University, Kocaeli, Turkey
| | - Wiebren Tjalma
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | | | - Glenn Broeckx
- Department of Histopathology, Antwerp University Hospital, Edegem, Belgium
| | - Karen Zwaenepoel
- Department of Histopathology, Antwerp University Hospital, Edegem, Belgium
| | - Maria Tholhuijsen
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Yannick Verhoeven
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Zwi Berneman
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium; Department of Hematology, Antwerp University, Edegem, Belgium
| | - Marc Peeters
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium; Department of Histopathology, Antwerp University Hospital, Edegem, Belgium
| | - Peter A van Dam
- Multidisciplinary Oncologic Centre Antwerp [(MOCA)], Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
36
|
Sanz-Ortega L, Rojas JM, Barber DF. Improving Tumor Retention of Effector Cells in Adoptive Cell Transfer Therapies by Magnetic Targeting. Pharmaceutics 2020; 12:E812. [PMID: 32867162 PMCID: PMC7557387 DOI: 10.3390/pharmaceutics12090812] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Adoptive cell transfer therapy is a promising anti-tumor immunotherapy in which effector immune cells are transferred to patients to treat tumors. However, one of its main limitations is the inefficient trafficking of inoculated effector cells to the tumor site and the small percentage of effector cells that remain activated when reaching the tumor. Multiple strategies have been attempted to improve the entry of effector cells into the tumor environment, often based on tumor types. It would be, however, interesting to develop a more general approach, to improve and facilitate the migration of specific activated effector lymphoid cells to any tumor type. We and others have recently demonstrated the potential for adoptive cell transfer therapy of the combined use of magnetic nanoparticle-loaded lymphoid effector cells together with the application of an external magnetic field to promote the accumulation and retention of lymphoid cells in specific body locations. The aim of this review is to summarize and highlight the recent findings in the field of magnetic accumulation and retention of effector cells in tumors after adoptive transfer, and to discuss the possibility of using this approach for tumor targeting with chimeric antigen receptor (CAR) T-cells.
Collapse
Affiliation(s)
- Laura Sanz-Ortega
- Center for Hematology and Regenerative Medicine (HERM), Department of Medicine, Karolinska Institute, 14183 Stockholm, Sweden;
| | - José Manuel Rojas
- Animal Health Research Centre (CISA)-INIA, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, 28130 Madrid, Spain;
| | - Domingo F. Barber
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, 28049 Madrid, Spain
| |
Collapse
|
37
|
Gomez-Macias GS, Molinar-Flores G, Lopez-Garcia CA, Santuario-Facio S, Decanini-Arcaute H, Valero-Elizondo J, Treviño-Alvarado V, Ortiz-Lopez R, Dono A, Esteban-Zubero E, Alatorre-Jimenez MA, Garza CV, Peña-Curiel O, Cardona-Huerta S. Immunotyping of tumor-infiltrating lymphocytes in triple-negative breast cancer and genetic characterization. Oncol Lett 2020; 20:140. [PMID: 32934708 PMCID: PMC7471657 DOI: 10.3892/ol.2020.12000] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) reflect the host immune response against cancer cells. Immunomodulators have been recently suggested as a novel therapeutic strategy against triple-negative breast cancer (TNBC). However, the TIL profile in TNBC has not been thoroughly investigated. In the present study, the percentage, immunophenotype and genetic profiles of TILs in pre-surgical tumor samples of patients with TNBC were evaluated prior to neoadjuvant chemotherapy (NAC). Patients diagnosed with breast cancer at Hospital San José TecSalud were consecutively and prospectively enrolled in the present study between August 2011 and August 2015. The pathological response to NAC was evaluated using the de Miller-Payne and MD Anderson Cancer Center system. TIL percentage (low, intermediate, and high) was evaluated using special hematoxylin-eosin staining on the core needle biopsies. The immunophenotype of TILs was assessed by immunohistochemistry (IHC) for CD3+, CD4+ and CD8+. In addition, the gene expression profile of CD3, CD4, CD8, CD20, CD45, forkhead box P3, interleukin 6, programmed cell death 1 and CD274 molecule was assessed in all patients. A total of 26 samples from patients with TNBC prior to NAC were included in the present study. TILs were low in 30.7%, intermediate in 38.4% and elevated in 30.7% of tumors. CD3+ and CD4+ counts were associated with the pathological response to NAC (P=0.04). Finally, an overexpression pattern of CD3, CD4, CD8, CD45 and CD20 genes was observed in patients with a partial or complete pathological response. The present results demonstrated that TILs may predict the pathological response to NAC in patients with TNBC. Furthermore, a more accurate association was identified between the high expression levels of CD3, CD4, CD8, CD45 and CD20 genes and partial and complete pathological response, compared with the association between high expression and IHC alone.
Collapse
Affiliation(s)
- Gabriela Sofia Gomez-Macias
- Department of Pathology, Hospital San José TecSalud, Tecnológico de Monterrey, Monterrey, Nuevo León 64710, México.,Department of Pathology, Hospital Universitario, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León 64460, México
| | - Guillermo Molinar-Flores
- Department of Pathology, Hospital San José TecSalud, Tecnológico de Monterrey, Monterrey, Nuevo León 64710, México
| | - Carlos A Lopez-Garcia
- Department of Pathology, Hospital San José TecSalud, Tecnológico de Monterrey, Monterrey, Nuevo León 64710, México
| | - Sandra Santuario-Facio
- Laboratorio de Investigacion, Tecnologico de Monterrey, Monterrey, Nuevo León 64710, México
| | - Horacio Decanini-Arcaute
- Department of Pathology, Hospital Christus Muguerza of Alta Especialidad, Monterrey, Nuevo León 64060, México
| | - Javier Valero-Elizondo
- Breast Cancer Center, Hospital San José, TecSalud, Tecnológico de Monterrey, Monterrey, Nuevo León 64710, México
| | | | - Rocio Ortiz-Lopez
- Laboratorio de Investigacion, Tecnologico de Monterrey, Monterrey, Nuevo León 64710, México
| | - Antonio Dono
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Medical School, Houston, TX 77030, USA
| | | | | | - Cynthia Villarreal Garza
- Breast Cancer Center, Hospital San José, TecSalud, Tecnológico de Monterrey, Monterrey, Nuevo León 64710, México
| | - Omar Peña-Curiel
- Breast Cancer Center, Hospital San José, TecSalud, Tecnológico de Monterrey, Monterrey, Nuevo León 64710, México
| | - Servando Cardona-Huerta
- Breast Cancer Center, Hospital San José, TecSalud, Tecnológico de Monterrey, Monterrey, Nuevo León 64710, México
| |
Collapse
|
38
|
Jo Y, Ali LA, Shim JA, Lee BH, Hong C. Innovative CAR-T Cell Therapy for Solid Tumor; Current Duel between CAR-T Spear and Tumor Shield. Cancers (Basel) 2020; 12:cancers12082087. [PMID: 32731404 PMCID: PMC7464778 DOI: 10.3390/cancers12082087] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Novel engineered T cells containing chimeric antigen receptors (CAR-T cells) that combine the benefits of antigen recognition and T cell response have been developed, and their effect in the anti-tumor immunotherapy of patients with relapsed/refractory leukemia has been dramatic. Thus, CAR-T cell immunotherapy is rapidly emerging as a new therapy. However, it has limitations that prevent consistency in therapeutic effects in solid tumors, which accounts for over 90% of all cancer patients. Here, we review the literature regarding various obstacles to CAR-T cell immunotherapy for solid tumors, including those that cause CAR-T cell dysfunction in the immunosuppressive tumor microenvironment, such as reactive oxygen species, pH, O2, immunosuppressive cells, cytokines, and metabolites, as well as those that impair cell trafficking into the tumor microenvironment. Next-generation CAR-T cell therapy is currently undergoing clinical trials to overcome these challenges. Therefore, novel approaches to address the challenges faced by CAR-T cell immunotherapy in solid tumors are also discussed here.
Collapse
Affiliation(s)
- Yuna Jo
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Laraib Amir Ali
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Ju A Shim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Byung Ha Lee
- NeoImmuneTech, Inc., 2400 Research Blvd., Suite 250, Rockville, MD 20850, USA;
| | - Changwan Hong
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
- Correspondence: ; Tel.: +82-51-510-8041
| |
Collapse
|
39
|
FoxP3 + T regulatory cells in cancer: Prognostic biomarkers and therapeutic targets. Cancer Lett 2020; 490:174-185. [PMID: 32721551 DOI: 10.1016/j.canlet.2020.07.022] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/28/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022]
Abstract
T Regulatory cells (Tregs) can have both protective and pathological roles. They maintain immune homeostasis and inhibit immune responses in various diseases, including cancer. Proportions of Tregs in the peripheral blood of some cancer patients increase by approximately two-fold, compared to those in healthy individuals. Tregs contribute to cancer development and progression by suppressing T effector cell functions, thereby compromising tumor killing and promoting tumor growth. Highly immunosuppressive Tregs express upregulated levels of the transcription factor, Forkhead box protein P3 (FoxP3). Elevated levels of FoxP3+ Tregs within the tumor microenvironment (TME) showed a positive correlation with poor prognosis in various cancer patients. Despite the success of immunotherapy, including the use of immune checkpoint inhibitors, a significant proportion of patients show low response rates as a result of primary or acquired resistance against therapy. Some of the mechanisms which underlie the development of therapy resistance are associated with Treg suppressive function. In this review, we describe Treg contribution to cancer development/progression, and the mechanisms of Treg-mediated immunosuppression. We discuss the prognostic significance of FoxP3+ Tregs in different cancers and their potential use as prognostic biomarkers. We also describe potential therapeutic strategies to target Tregs in combination with other types of immunotherapies aiming to overcome tumor resistance and improve clinical outcomes in cancer patients. Overall, understanding the prognostic significance of FoxP3+ Tregs in various cancers and their contribution to therapy resistance could help in the development of more effective targeted therapeutic strategies to enhance the clinical outcomes in cancer patients.
Collapse
|
40
|
Simon B, Uslu U. Fasten the seat belt: Increasing safety of CAR T-cell therapy. Exp Dermatol 2020; 29:1039-1045. [PMID: 32627228 DOI: 10.1111/exd.14131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/01/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
After the recent success and approvals of chimeric antigen receptor (CAR) T cells in haematological malignancies, its efficacy is currently evaluated in a broad spectrum of tumor entities including melanoma. However, severe and potentially life-threatening side effects like cytokine release syndrome, neurologic toxicities, and the competing risk of morbidity and mortality from the treatment itself are still a major limiting factor in the current CAR T-cell landscape. In addition, especially in solid tumors, the lack of ideal target antigens to avoid on-target/off-tumor toxicities also restricts its use. While various groups are working on strategies to boost CAR T-cell efficacy, mechanisms to increase engineered T-cell safety should not move out of focus. Thus, the aim of this article is to summarize and to discuss current and potential future strategies and mechanisms to increase CAR T-cell safety in order to enable the wide use of this promising approach in melanoma and other tumor entities.
Collapse
Affiliation(s)
- Bianca Simon
- Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany.,Comprehensive Cancer Center Erlangen- European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.,Division of Genetics, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ugur Uslu
- Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany.,Comprehensive Cancer Center Erlangen- European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
41
|
Levels of different subtypes of tumour-infiltrating lymphocytes correlate with each other, with matched circulating lymphocytes, and with survival in breast cancer. Breast Cancer Res Treat 2020; 183:49-59. [PMID: 32577938 PMCID: PMC7376517 DOI: 10.1007/s10549-020-05757-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/16/2020] [Indexed: 02/08/2023]
Abstract
Purpose Breast cancer tumour-infiltrating lymphocytes associate with clinico-pathological factors, including survival, although the literature includes many conflicting findings. Our aim was to assess these associations for key lymphocyte subtypes and in different tumour compartments, to determine whether these provide differential correlations and could, therefore, explain published inconsistencies. Uniquely, we also examine whether infiltrating levels merely reflect systemic lymphocyte levels or whether local factors are predominant in recruitment. Methods Immunohistochemistry was used to detect tumour-infiltrating CD20+ (B), CD4+ (helper T), CD8+ (cytotoxic T) and FoxP3+ (regulatory T) cells in breast cancers from 62 patients, with quantification in tumour stroma, tumour cell nests, and tumour margins. Levels were analysed with respect to clinico-pathological characteristics and matched circulating levels (determined by flow-cytometry). Results CD4+ lymphocytes were the most prevalent subtype in tumour stroma and at tumour edge and CD8+ lymphocytes were most prevalent in tumour nests; FoxP3+ lymphocytes were rarest in all compartments. High grade or hormone receptor negative tumours generally had significantly increased lymphocytes, especially in tumour stroma. Only intra-tumoural levels of CD8+ lymphocytes correlated significantly with matched circulating levels (p < 0.03), suggesting that recruitment is mainly unrelated to systemic activity. High levels of stromal CD4+ and CD20+ cells associated with improved survival in hormone receptor negative cases (p < 0.04), while tumour nest CD8+ and FoxP3+ cells associated with poor survival in hormone receptor positives (p < 0.005). Conclusions Lymphocyte subtype and location define differential impacts on tumour biology, therefore, roles of tumour-infiltrating lymphocytes will only be unravelled through thorough analyses that take this into account. Electronic supplementary material The online version of this article (10.1007/s10549-020-05757-5) contains supplementary material, which is available to authorized users.
Collapse
|
42
|
Clinicopathologic Characteristics of Breast Cancer According to the Infiltrating Immune Cell Subtypes. Int J Mol Sci 2020; 21:ijms21124438. [PMID: 32580398 PMCID: PMC7352832 DOI: 10.3390/ijms21124438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/31/2022] Open
Abstract
The clinical significance of immune cell subtypes in breast cancer remains poorly understood. To identify tumor-infiltrating immune cell subtypes in breast cancer and investigate their implications, tissue microarrays were constructed using 334 cases of invasive ductal carcinoma (luminal A type: 162 (48.5%), luminal B type: 96 (28.7%), HER-2 type: 21 (6.3%), and triple negative breast cancer: 55 (16.5%)). Hormone receptors (ER, PR, and HER-2), Ki-67, and immune cell subtype-related proteins (STAT4, STAT6, FOXP3, CD8, CD68, and CD163) were assessed immunohistochemically. The proportion of highly expressed STAT6, FOXP3, CD8, CD68, and CD163 proteins was found to be lowest in luminal A type but highest in the HER-2 type. Additionally, high-level STAT6, FOXP3, CD68, and CD163 protein expression was associated with higher histologic grade. ER negativity was associated with high STAT6, FOXP3, and CD163 expression levels, whereas PR negativity and high Ki-67 labeling index were associated with high CD163 expression. Univariate (p = 0.003) and multivariate Cox (hazard ratio: 2.435, 95% CI: 1.110-5.344, p = 0.049) analyses showed that high CD8 expression is an independent factor associated with shorter disease-free survival. Immune cell subtype-related protein expression is dependent on breast cancer molecular subtypes, and CD8 expression is associated with patient prognosis.
Collapse
|
43
|
Bai F, Zhang P, Fu Y, Chen H, Zhang M, Huang Q, Li D, Li B, Wu K. Targeting ANXA1 abrogates Treg-mediated immune suppression in triple-negative breast cancer. J Immunother Cancer 2020; 8:e000169. [PMID: 32300050 PMCID: PMC7204868 DOI: 10.1136/jitc-2019-000169] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Regulatory T (Treg) cells play a negative role in anti-tumor immunity against triple-negative breast cancer, so it is of great significance to find the potential therapeutic target of Treg cells. METHODS First, Annexin A1 (ANXA1) expression and survival of patients with breast cancer were analyzed using TCGA data. Then plasma ANXA1 levels in patients with malignant and benign breast tumors were detected by ELISA. Next, the effect of ANXA1 on Treg cells was studied through suppressive assays, and how ANXA1 regulates the function of Treg cells was detected by RNA sequencing. Finally, the in vivo experiment in balb/c mice was conducted to test whether the ANXA1 blocker Boc1 could shrink tumors and affect the function of Treg cells. RESULTS Our data suggest that ANXA1 expression is associated with lower survival and a higher risk of breast malignancy. Suppressive assays show that ANXA1 can enhance the inhibition function of Treg cells. RNA-Sequencing results indicate that Boc1 could reduce the expression of granzyme A mRNA in Treg cells. Animal experiments have been done to show that Boc1 can reduce tumor size and down regulate Treg cell function. CONCLUSIONS ANXA1 can enhance the function of Treg cells and reduce the survival rate of patients with breast cancer. Targeting ANXA1 can reduce Treg cell function and shrink breast tumors.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Annexin A1/antagonists & inhibitors
- Annexin A1/genetics
- Annexin A1/metabolism
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Lobular/immunology
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/pathology
- Cell Movement
- Cell Proliferation
- Female
- Follow-Up Studies
- Gene Expression Regulation, Neoplastic
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Prognosis
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Survival Rate
- T-Lymphocytes, Regulatory/immunology
- Triple Negative Breast Neoplasms/immunology
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Fang Bai
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Peng Zhang
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yipeng Fu
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Hongliang Chen
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Mingdi Zhang
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Qianru Huang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kejin Wu
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
44
|
Solis-Castillo LA, Garcia-Romo GS, Diaz-Rodriguez A, Reyes-Hernandez D, Tellez-Rivera E, Rosales-Garcia VH, Mendez-Cruz AR, Jimenez-Flores JR, Villafana-Vazquez VH, Pedroza-Gonzalez A. Tumor-infiltrating regulatory T cells, CD8/Treg ratio, and cancer stem cells are correlated with lymph node metastasis in patients with early breast cancer. Breast Cancer 2020; 27:837-849. [PMID: 32180141 DOI: 10.1007/s12282-020-01079-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/05/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Tumor-infiltrating lymphocytes are an important component of the tumor microenvironment (TME) in breast cancer. They have been linked with tumor pathogenesis in advanced stages. However, little is known about their contribution in early phases. In this study, we analyzed the infiltration of leukocytes and cancer stem cells (CSC) in tumors from patients with early breast cancer. METHODS Samples of blood and tumor tissue from 30 patients with breast cancer were collected, and the number of dendritic cells (DC), T cells, and CSC were analyzed by flow cytometry. RESULTS Tumor-infiltrating CD4 and CD8 T cells expressed higher levels of cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) compared with peripheral T cells. Regulatory T cells (Treg) were enriched in tumors and overexpressed glucocorticoid-induced TNFR-related protein and CTLA-4. Tumor Treg had a positive correlation with the amount of myeloid DC (mDC) and disease progression. The CD8/Treg ratio was associated with lymph node metastasis and tumor stages. The main subset of DC in early breast tumors was mDC, while plasmacytoid DC were almost absent. CSC were present in most tumors with higher frequencies in patients with lymph node metastasis. CSC were also associated with the amount of tumor-infiltrating Treg. CONCLUSION Early breast cancer has an inflammatory milieu characterized by mDC, Treg, and CSC infiltration. The frequencies of Treg, CSC and CD8/Treg ratio were associated with disease progression. The composition of leukocytes and the presence of CSC in early breast tumors should be considered for the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Luis Alberto Solis-Castillo
- Hospital de Gineco Obstetricia No. 3 del Centro Médico Nacional la Raza del Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Gina Stella Garcia-Romo
- Unidad de morfología y función (UMF), Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Ave. De los Barrios No. 1., 54090, Tlalnepantla, Estado de México, México.,Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Estado de México, México
| | - Alvaro Diaz-Rodriguez
- Unidad de morfología y función (UMF), Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Ave. De los Barrios No. 1., 54090, Tlalnepantla, Estado de México, México
| | - Diana Reyes-Hernandez
- Unidad de morfología y función (UMF), Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Ave. De los Barrios No. 1., 54090, Tlalnepantla, Estado de México, México
| | - Elizabeth Tellez-Rivera
- Unidad de morfología y función (UMF), Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Ave. De los Barrios No. 1., 54090, Tlalnepantla, Estado de México, México
| | - Victor Hugo Rosales-Garcia
- Laboratorios Nacionales de Servicios Experimentales (LANSE), Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, México
| | - Adolfo Rene Mendez-Cruz
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Estado de México, México
| | - Jose Rafael Jimenez-Flores
- Unidad de morfología y función (UMF), Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Ave. De los Barrios No. 1., 54090, Tlalnepantla, Estado de México, México.,Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Estado de México, México
| | - Victor Hugo Villafana-Vazquez
- Hospital de Gineco Obstetricia No. 3 del Centro Médico Nacional la Raza del Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Alexander Pedroza-Gonzalez
- Unidad de morfología y función (UMF), Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Ave. De los Barrios No. 1., 54090, Tlalnepantla, Estado de México, México. .,Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Estado de México, México. .,Unidad de investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores Iztacala de la Universidad Nacional Autónoma de México, Estado de México, México.
| |
Collapse
|
45
|
Moody R, Wilson K, Jaworowski A, Plebanski M. Natural Compounds with Potential to Modulate Cancer Therapies and Self-Reactive Immune Cells. Cancers (Basel) 2020; 12:cancers12030673. [PMID: 32183059 PMCID: PMC7139800 DOI: 10.3390/cancers12030673] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer-related deaths are approaching 10 million each year. Survival statistics for some cancers, such as ovarian cancer, have remained unchanged for decades, with women diagnosed at stage III or IV having over 80% chance of a lethal cancer recurrence after standard first-line treatment (reductive surgery and chemotherapy). New treatments and adjunct therapies are needed. In ovarian cancer, as in other cancers, the immune response, particularly cytotoxic (CD8+) T cells are correlated with a decreased risk of recurrence. As well as completely new antigen targets resulting from DNA mutations (neo-antigens), these T cells recognize cancer-associated overexpressed, re-expressed or modified self-proteins. However, there is concern that activation of self-reactive responses may also promote off-target pathology. This review considers the complex interplay between cancer-reactive and self-reactive immune cells and discusses the potential uses for various leading immunomodulatory compounds, derived from plant-based sources, as a cancer therapy option or to modulate potential autoimmune pathology. Along with reviewing well-studied compounds such as curcumin (from turmeric), epigallocatechin gallate (EGCG, from green tea) and resveratrol (from grapes and certain berries), it is proposed that compounds from novel sources, for example, native Australian plants, will provide a useful source for the fine modulation of cancer immunity in patients.
Collapse
|
46
|
Higher density of stromal M2 macrophages in breast ductal carcinoma in situ predicts recurrence. Virchows Arch 2020; 476:825-833. [PMID: 31897820 DOI: 10.1007/s00428-019-02735-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 10/25/2022]
Abstract
Immune response can affect tumour progression and treatment outcome. This study investigated the potential of stromal macrophages around ductal carcinoma in situ (DCIS) in predicting recurrence and progression. CD68 and CD163 expression of macrophages in DCIS from 198 patients was determined by immunohistochemistry. Disease free survival (DFS), clinicopathological parameters and biomarker expression were correlated with the densities of both CD68+ and CD163+ macrophages. High CD68+ macrophage density was associated with high nuclear grade (p < 0.001), oestrogen receptor (ER) negativity (p = 0.029), progesterone receptor (PR) negativity (p = 0.008) and human epidermal growth factor receptor 2 (HER2) positivity (p < 0.001). High CD163+ macrophage density was associated with high nuclear grade (p = 0.003), microinvasion (p = 0.01), ER negativity (p < 0.001), PR negativity (p = 0.001), HER2 positivity (p = 0.001) and triple negativity (p = 0.022). DCIS with higher CD68+ macrophage density disclosed significantly worse DFS for ipsilateral invasive recurrence (p = 0.004) and is affirmed by multivariate Cox regression analysis (95% CI 1.126-5.102, HR = 2.397, p = 0.023). DCIS with higher CD163+ macrophage density showed significantly worse DFS for both recurrence (p = 0.001) and ipsilateral invasive recurrence (p = 0.001). These findings, for CD163+ macrophage density, were affirmed by multivariate Cox regression analysis respectively for both recurrence (95% CI 1.210-2.293, HR = 1.880, p = 0.005) and ipsilateral invasive recurrence (95% CI 1.122-5.176, HR = 2.410, p = 0.024). This study demonstrated that DCIS with higher macrophage density was associated with poorer prognostic parameters, while DCIS with higher CD163+ macrophage density predicted both recurrence and ipsilateral invasive recurrence.
Collapse
|
47
|
Hashemi V, Maleki LA, Esmaily M, Masjedi A, Ghalamfarsa G, Namdar A, Yousefi M, Yousefi B, Jadidi-Niaragh F. Regulatory T cells in breast cancer as a potent anti-cancer therapeutic target. Int Immunopharmacol 2019; 78:106087. [PMID: 31841758 DOI: 10.1016/j.intimp.2019.106087] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/23/2019] [Accepted: 11/25/2019] [Indexed: 02/08/2023]
Abstract
Despite marked advances in treatment approaches, breast cancer is still going to be more prevalent, worldwide. High levels of regulatory T (Treg) cells have repeatedly been demonstrated in circulation, lymph nodes, and tumor samples from patients with various cancer types. The transcription factor Forkhead box protein 3 (Foxp3)-expressing Treg cells have the high suppressive potential of the immune system and are fundamental in preserving immune homeostasis and self-tolerance. However, they enhance tumor development by curbing efficient anti-tumor immune mechanisms in malignancies. Moreover, the accumulation of Treg cells in breast tumors is related to the short overall survival of patients. Treg cell frequency has been applied as an independent predicting factor to diagnose patients with a high risk of relapse. Pulling out all populations of Treg cells to promote the efficacy of anticancer treatment methods may potentially lead to hazardous autoimmune disorders. Thus, realizing the exact structure of tumor-infiltrating Treg cells is pivotal to efficiently target Treg cells in tumors. There are exclusive and non-exclusive approaches to lower down and degrade the number/function of Treg cells. These approaches can include inhibiting tumoral migration, depletion, interference with function, and utilizing T cell plasticity. This review article attempts to clarify the implications concerning the involvement of Treg cells in breast cancer progression and discuss the current approaches in the treatment of this cancer via modulation of Treg cells function.
Collapse
Affiliation(s)
- Vida Hashemi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Maryam Esmaily
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Masjedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Afshin Namdar
- Katz Group Centre for Pharmacy and Health Research, University of Alberta, Edmonton, Canada
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
48
|
Lesch S, Benmebarek MR, Cadilha BL, Stoiber S, Subklewe M, Endres S, Kobold S. Determinants of response and resistance to CAR T cell therapy. Semin Cancer Biol 2019; 65:80-90. [PMID: 31705998 DOI: 10.1016/j.semcancer.2019.11.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/28/2019] [Accepted: 11/03/2019] [Indexed: 12/27/2022]
Abstract
The remarkable success of chimeric antigen receptor (CAR)-engineered T cells in pre-B cell acute lymphoblastic leukemia (ALL) and B cell lymphoma led to the approval of anti-CD19 CAR T cells as the first ever CAR T cell therapy in 2017. However, with the number of CAR T cell-treated patients increasing, observations of tumor escape and resistance to CAR T cell therapy with disease relapse are demonstrating the current limitations of this therapeutic modality. Mechanisms hampering CAR T cell efficiency include limited T cell persistence, caused for example by T cell exhaustion and activation-induced cell death (AICD), as well as therapy-related toxicity. Furthermore, the physical properties, antigen heterogeneity and immunosuppressive capacities of solid tumors have prevented the success of CAR T cells in these entities. Herein we review current obstacles of CAR T cell therapy and propose strategies in order to overcome these hurdles and expand CAR T cell therapy to a broader range of cancer patients.
Collapse
Affiliation(s)
- Stefanie Lesch
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Bruno L Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Stefan Stoiber
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Marion Subklewe
- German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany; Department of Medicine III, Klinikum der Universität München, LMU Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany; German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany; German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany.
| |
Collapse
|
49
|
Simon B, Uslu U. CAR-T cell therapy in melanoma: A future success story? Exp Dermatol 2019; 27:1315-1321. [PMID: 30288790 DOI: 10.1111/exd.13792] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/23/2018] [Accepted: 09/26/2018] [Indexed: 12/13/2022]
Abstract
Chimeric antigen receptor (CAR)-T cells are one of the impressive recent success stories of anti-cancer immunotherapy. Especially in haematological malignancies, this treatment strategy has shown promising results leading to the recent approval of two CAR-T cell constructs targeting CD19 in the United States and the European Union. After the huge success in haematological cancers, the next step will be the evaluation of its efficacy in different solid tumors, which is currently investigated in preclinical as well as clinical settings. A commonly examined tumor model in the context of immunotherapy is melanoma, since it is known for its immunogenic features. However, the first results of CAR-T cell therapy in solid tumors did not reveal the same impressive outcomes that were observed in haematological malignancies, as engineered cells need to cope with several challenges. Obstacles include the lack of migration of CAR-T cells from blood vessels to the tumor site as well as the immunosuppressive tumor microenvironment within solid tumors. Another hurdle is posed by the identification of an ideal target antigen to avoid on-target/off-tumor toxicities. Regarding immune escape mechanisms, which can be developed by tumor cells to bypass immune recognition, the observation of antigen loss should also be considered. This article gives an overview of the challenges displayed in CAR-T cell therapy for the use in solid tumors and discusses different new strategies and approaches that deal with these problems in order to improve CAR-T cell therapy, particularly for its use in melanoma.
Collapse
Affiliation(s)
- Bianca Simon
- Department of Dermatology, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Biology, Division of Genetics, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ugur Uslu
- Department of Dermatology, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
50
|
Sanz-Ortega L, Portilla Y, Pérez-Yagüe S, Barber DF. Magnetic targeting of adoptively transferred tumour-specific nanoparticle-loaded CD8 + T cells does not improve their tumour infiltration in a mouse model of cancer but promotes the retention of these cells in tumour-draining lymph nodes. J Nanobiotechnology 2019; 17:87. [PMID: 31387604 PMCID: PMC6683429 DOI: 10.1186/s12951-019-0520-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/30/2019] [Indexed: 12/16/2022] Open
Abstract
Background Adoptive T cell-transfer (ATC) therapy is a highly promising cancer-treatment approach. However, in vivo-administered T cells tend to disperse, with only a small proportion reaching the tumour. To remedy this, magnetic targeting of T cells has been recently explored. Magnetic nanoparticles (MNPs) functionalised with antibodies were attached to effector T cells and magnetically recruited to tumour sites under MRI guidance. In this study, we investigated whether 3-aminopropyl-triethoxysilane (APS)-coated MNPs directly attached to CD8+ T cell membranes could also magnetically target and accumulate tumour-specific CD8+ T cells in solid tumours using an external magnetic field (EMF). As it has been shown that T cells associated with APS-coated MNPs are retained in lymph nodes (LNs), and tumour-draining LNs are the most common sites of solid-tumour metastases, we further evaluated whether magnetic targeting of APS-MNP-loaded CD8+ T cells could cause them to accumulate in tumour-draining LNs. Results First, we show that antigen-specific CD8+ T cells preserve their antitumor activity in vitro when associated with APS-MNPs. Next, we demonstrate that the application of a magnetic field enhanced the retention of APS-MNP-loaded OT-I CD8+ T cells under flow conditions in vitro. Using a syngeneic mouse model, we found similar numbers of APS-MNP-loaded OT-I CD8+ T cells and OT-I CD8+ T cells infiltrating the tumour 14 days after cell transfer. However, when a magnet was placed near the tumour during the transfer of tumour-specific APS-MNP-loaded CD8+ T cells to improve tumour infiltration, a reduced percentage of tumour-specific T cells was found infiltrating the tumour 14 days after cell transfer, which was reflected in a smaller reduction in tumour size compared to tumour-specific CD8+ T cells transferred with or without MNPs in the absence of a magnetic field. Nonetheless, magnet placement near the tumour site during cell transfer induced infiltration of activated tumour-specific CD8+ T cells in tumour-draining LNs, which remained 14 days after cell transfer. Conclusions The use of an EMF to improve targeting of tumour-specific T cells modified with APS-MNPs reduced the percentage of these cells infiltrating the tumour, but promoted the retention and the persistence of these cells in the tumour-draining LNs. ![]() Electronic supplementary material The online version of this article (10.1186/s12951-019-0520-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura Sanz-Ortega
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Darwin 3, Cantoblanco, 28049, Madrid, Spain
| | - Yadileiny Portilla
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Darwin 3, Cantoblanco, 28049, Madrid, Spain
| | - Sonia Pérez-Yagüe
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Darwin 3, Cantoblanco, 28049, Madrid, Spain
| | - Domingo F Barber
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Darwin 3, Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|