51
|
Kerneur C, Cano CE, Olive D. Major pathways involved in macrophage polarization in cancer. Front Immunol 2022; 13:1026954. [PMID: 36325334 PMCID: PMC9618889 DOI: 10.3389/fimmu.2022.1026954] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages play an important role in tissue homeostasis, tissue remodeling, immune response, and progression of cancer. Consequently, macrophages exhibit significant plasticity and change their transcriptional profile and function in response to environmental, tissue, and inflammatory stimuli resulting in pro- and anti-tumor effects. Furthermore, the categorization of tissue macrophages in inflammatory situations remains difficult; however, there is an agreement that macrophages are predominantly polarized into two different subtypes with pro- and anti-inflammatory properties, the so-called M1-like and M2-like macrophages, respectively. These two macrophage classes can be considered as the extreme borders of a continuum of many intermediate subsets. On one end, M1 are pro-inflammatory macrophages that initiate an immunological response, damage tissue integrity, and dampen tumor progression by fostering robust T and natural killer (NK) cell anti-tumoral responses. On the other end, M2 are anti-inflammatory macrophages involved in tissue remodeling and tumor growth, that promote cancer cell proliferation, invasion, tumor metastasis, angiogenesis and that participate to immune suppression. These decisive roles in tumor progression occur through the secretion of cytokines, chemokines, growth factors, and matrix metalloproteases, as well as by the expression of immune checkpoint receptors in the case of M2 macrophages. Moreover, macrophage plasticity is supported by stimuli from the Tumor Microenvironment (TME) that are relayed to the nucleus through membrane receptors and signaling pathways that result in gene expression reprogramming in macrophages, thus giving rise to different macrophage polarization outcomes. In this review, we will focus on the main signaling pathways involved in macrophage polarization that are activated upon ligand-receptor recognition and in the presence of other immunomodulatory molecules in cancer.
Collapse
Affiliation(s)
- Clément Kerneur
- ImCheck Therapeutics, Marseille, France
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, CNRS UMR7258, Institut Paoli Calmettes, Marseille, France
- *Correspondence: Clément Kerneur, ; Carla E. Cano, ; Daniel Olive,
| | - Carla E. Cano
- ImCheck Therapeutics, Marseille, France
- *Correspondence: Clément Kerneur, ; Carla E. Cano, ; Daniel Olive,
| | - Daniel Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, CNRS UMR7258, Institut Paoli Calmettes, Marseille, France
- *Correspondence: Clément Kerneur, ; Carla E. Cano, ; Daniel Olive,
| |
Collapse
|
52
|
Dhandapani H, Bose M, Kesavan S. The Immune-related ceRNA Network in Prognosis of Cervical Cancer. Asian Pac J Cancer Prev 2022; 23:3347-3354. [PMID: 36308358 PMCID: PMC9924325 DOI: 10.31557/apjcp.2022.23.10.3347] [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: 03/04/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Immunotherapy is gaining attention and it is being included as one of the treatment strategies for cancer patients. However, the molecular mechanisms of immune-related genes and their affinity for cervical cancer progression remain unclear. In this study, we have developed an immune-related competing endogenous RNA [ceRNA] network and assessed the tumour infiltrating immune cells towards the prognosis of cervical cancer. METHODS Differential RNA expression pattern between stages I and II-IV of cervical cancer patients from The Cancer Genome Atlas [TCGA] was analyzed. Immune-related ceRNA network based on the immune gene signatures were retrieved and their targets were predicted using miRwalk 3.0. CIBERSORT was employed to identify the immune cell types based on their respective transcripts. The prognostic significance of RNAs in the ceRNA network and immune cell subsets was analyzed. RESULTS Significant differences in 22 long non-coding RNAs [lncRNAs], 15 microRNAs [miRNAs], and 252 messenger RNAs [mRNAs] between stages I and II-IV of cervical cancer were observed. Further, we shortlisted the 49 immune-related mRNAs based on immune gene signature and predicted their target miRNAs and lncRNAs. A potential ceRNA network of 4 lncRNAs, 10 miRNAs, and 11 mRNAs had a strong correlation for prognosis. Out of 11 protein-coding immune mRNAs, IRF4 and AZGP1 had high degrees of interaction. In addition, the evaluation of immune cell subsets showed increased infiltration of M1 macrophages had better survival outcome. CONCLUSIONS We have identified an immune-related ceRNA network based on differentially expressed transcripts between stages I and II-IV which may help predict the prognosis of cervical cancer.
Collapse
|
53
|
Tang WW, Bauer KM, Barba C, Ekiz HA, O’Connell RM. miR-aculous new avenues for cancer immunotherapy. Front Immunol 2022; 13:929677. [PMID: 36248881 PMCID: PMC9554277 DOI: 10.3389/fimmu.2022.929677] [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: 04/27/2022] [Accepted: 08/18/2022] [Indexed: 01/25/2023] Open
Abstract
The rising toll of cancer globally necessitates ingenuity in early detection and therapy. In the last decade, the utilization of immune signatures and immune-based therapies has made significant progress in the clinic; however, clinical standards leave many current and future patients without options. Non-coding RNAs, specifically microRNAs, have been explored in pre-clinical contexts with tremendous success. MicroRNAs play indispensable roles in programming the interactions between immune and cancer cells, many of which are current or potential immunotherapy targets. MicroRNAs mechanistically control a network of target genes that can alter immune and cancer cell biology. These insights provide us with opportunities and tools that may complement and improve immunotherapies. In this review, we discuss immune and cancer cell-derived miRNAs that regulate cancer immunity and examine miRNAs as an integral part of cancer diagnosis, classification, and therapy.
Collapse
Affiliation(s)
- William W. Tang
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Kaylyn M. Bauer
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Cindy Barba
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Huseyin Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, İzmir, Turkey
| | - Ryan M. O’Connell
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
54
|
Chabeli MS, Wang X, Yinghao L, Chen C, Yang C, Shou Y, Wang S, Chen K. Similarities between wound re-epithelialization and Metastasis in ESCC and the crucial involvement of macrophages: A review. Cancer Treat Res Commun 2022; 32:100621. [PMID: 36007473 DOI: 10.1016/j.ctarc.2022.100621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
In cancer, tumor-associated macrophages (TAMs) possess crucial functions in facilitating epithelial-mesenchymal transition (EMT). EMT is a crucial process in tumor metastasis. Tumor metastasis is one of the hallmarks of cancer and leads to patient mortality. Cancer cells often find ways to evade being detected and attacked by the immune system. This is achieved by cross-talk between cancer cells and the altered microenvironment. The accumulation of tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) creates an immunosuppressive and tumor-supportive environment. Circulating monocytes and macrophages which are recruited into tumors are defined as tumor-associated macrophages once in the TME. Based on the activated stimuli and function, macrophages can be divided into M1 macrophages and M2 macrophages. M1 macrophages, also known as classically activated macrophages, exhibit pro-inflammatory and antitumor activities. M2 macrophages, also known as alternatively activated macrophages, exhibit anti-inflammatory, pro-tumorigenic, and wound healing activities. TAMs are considered to be of the M2 phenotype. The TME polarizes recruited macrophages into M2 macrophages as they provide an immunosuppressive pro-tumoral environment. Accumulating studies show that the presence of TAMs in esophageal squamous cell carcinoma (ESCC) leads to tumor progression. In this review, we discuss how EMT can be used by TAMs to cause tumor migration and metastasis in ESCC. We also discuss the potential therapies targeting TAMs.
Collapse
Affiliation(s)
- Maletsooa Story Chabeli
- Academy of medical sciences, Department of Pathology, Zhengzhou University, Zhengzhou, Henan, China; Provincial Key Laboratory of Tumor Pathology, Zhengzhou, 450052, China,.
| | - Xiaoqian Wang
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Liang Yinghao
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chao Chen
- Academy of medical sciences, Department of Pathology, Zhengzhou University, Zhengzhou, Henan, China; Provincial Key Laboratory of Tumor Pathology, Zhengzhou, 450052, China
| | - Chenbo Yang
- Provincial Key Laboratory of Tumor Pathology, Zhengzhou, 450052, China
| | - Yuwei Shou
- Academy of medical sciences, Department of Pathology, Zhengzhou University, Zhengzhou, Henan, China; Provincial Key Laboratory of Tumor Pathology, Zhengzhou, 450052, China
| | - Shuaiyuan Wang
- Provincial Key Laboratory of Tumor Pathology, Zhengzhou, 450052, China
| | - Kuisheng Chen
- Academy of medical sciences, Department of Pathology, Zhengzhou University, Zhengzhou, Henan, China; Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Henan, China; Provincial Key Laboratory of Tumor Pathology, Zhengzhou, 450052, China,.
| |
Collapse
|
55
|
An Immune-Related lncRNA Pairing Model for Predicting the Prognosis and Immune-Infiltrating Cell Condition in Human Ovarian Cancer. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3168408. [PMID: 36033566 PMCID: PMC9400430 DOI: 10.1155/2022/3168408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/20/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022]
Abstract
Ovarian cancer is the second common cancer among the gynecological tumors. It is difficult to be found and diagnosed in the early stage and easy to relapse due to chemoresistance and deficiency in choices of treatment. Therefore, future exploring the biomarkers for diagnosis, treatment, and prognosis prediction of ovarian cancer is significant to women in the world. We downloaded data from TCGA and GTEx and used R “limma” package for analyzing the differentially expressed immune-related lncRNA in ovarian cancer and finally got 7 downregulated and 171 upregulated lncRNA. Then, we paired the differentially expressed immune-related lncRNA and constructed a novel lncRNA pairing model containing 7 lncRNA pairs. Based on the cut-off point with the highest AUC value, 102 patients were selected in high-risk group and 272 in low-risk group. The KM analysis suggested that the patients in the low-risk group had a longer overall survival. Future analysis showed the correlations between risk scores and clinicopathological parameters and infiltrating immune cells. In conclusion, we identified an immune-related lncRNA pairing model for predicting the prognosis and immune-infiltrating cell condition in human ovarian cancer, which thus further can instruct immunotherapy.
Collapse
|
56
|
Wang H, Liu J, Yang J, Wang Z, Zhang Z, Peng J, Wang Y, Hong L. A novel tumor mutational burden-based risk model predicts prognosis and correlates with immune infiltration in ovarian cancer. Front Immunol 2022; 13:943389. [PMID: 36003381 PMCID: PMC9393426 DOI: 10.3389/fimmu.2022.943389] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022] Open
Abstract
Tumor mutational burden (TMB) has been reported to determine the response to immunotherapy, thus affecting the patient’s prognosis in many cancers. However, it is unclear whether TMB or TMB-related signature could be used as prognostic indicators for ovarian cancer (OC), as its potential association with immune infiltration remains poorly understood. Therefore, this study aimed to develop a novel TMB-related risk model (TMBrisk) to predict the prognosis of OC patients on the basis of exploring TMB-related genes, and to explore the potential association between TMB/TMBrisk and immune infiltration. The mutational landscape, TMB scores, and correlations between TMB and clinical characteristics and immune infiltration were investigated in The Cancer Genome Atlas (TCGA)-OV cohort. Differentially expressed gene (DEG) analyses and weighted gene co-expression network analysis (WGCNA) were performed to derive TMB-related genes. TMBrisk was constructed by Cox regression and further validated in Gene Expression Omnibus (GEO) datasets. The mRNA and protein expression levels and biological functions of TMBrisk hub genes were verified through Gene Expression Profiling Interactive Analysis (GEPIA), GSCA Lite, the Human Protein Atlas (HPA) database, and RT-qPCR. TMBrisk-related biological phenotypes were analyzed in function enrichment and tumor immune infiltration signature. Potential therapeutic regimens were inferred utilizing the Genomics of Drug Sensitivity in Cancer (GDSC) database and connectivity map (CMap). According to our results, higher TMB was associated with better survival and higher CD8+ T cell, regulatory T cell, and NK cell infiltration. TMBrisk was developed based on CBWD1, ST7L, RFX5-AS1, C3orf38, LRFN1, LEMD1, and HMGB1. High TMBrisk was identified as a poor factor for prognosis in TCGA and GEO datasets; the high-TMBrisk group comprised more higher-grade (G2 and G3) and advanced clinical stage (stage III/IV) tumors. Meanwhile, higher TMBrisk was associated with an immunosuppressive phenotype, with less infiltration of a majority of immunocytes and less expression of several genes of the human leukocyte antigen (HLA) family. Moreover, a nomogram containing TMBrisk showed a strong predictive ability demonstrated by time-dependent ROC analysis. Overall, this novel TMB-related risk model (TMBrisk) could predict prognosis, evaluate immune infiltration, and discover new therapeutic regimens in OC, which is very promising in clinical promotion.
Collapse
|
57
|
Wang S, Wang X, Xia X, Zhang T, Yi M, Li Z, Jiang L, Yang Y, Fu J, Fang X. Identification of the immune subtype of ovarian cancer patients by integrated analyses of transcriptome and single-cell sequencing data. Sci Rep 2022; 12:13296. [PMID: 35918500 PMCID: PMC9346122 DOI: 10.1038/s41598-022-17645-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/28/2022] [Indexed: 11/09/2022] Open
Abstract
Ovarian cancer (OC) is one the most life-threatening cancers affecting women's health worldwide. Immunotherapy has become a promising treatment for a variety of cancers, but the therapeutic effects in OC remain limited. In this study, we constructed a macrophage risk score (MRS) based on M1 and M2 macrophages and a gene risk score (GRS) based on the prognostic genes associated with MRS. Next, cell-cell communication analysis was performed using single-cell RNA (scRNA) sequencing data. Survival status and immune characteristics were compared between the high- and low-score groups separated by MRS or GRS. Our results suggested that MRS and GRS can identify the immune subtypes of OC patients with better overall survival (OS) and inflammatory immune microenvironment. Moreover, M1 and M2 macrophages may affect the prognosis of OC patients through signal communication with CD8 T cells. Finally, functional differences between the two groups separated by GRS were elucidated. Taken together, this study constructed two useful models for the identification of immune subtypes in OC, which has a better prognosis and may have a sensitive response to immune checkpoint inhibitors (ICIs). The hub genes for the construction of GRS may be potential synergetic targets for immunotherapy in OC patients.
Collapse
Affiliation(s)
- Sixue Wang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xi Wang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaomeng Xia
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Tingting Zhang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Mingyu Yi
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zeying Li
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Jiang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yalan Yang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jie Fu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Xiaoling Fang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
58
|
Feng Y, Xiao M, Cao G, Liu H, Li Y, Wang S, Zijtveld S, Delvoux B, Xanthoulea S, Romano A, Liu C, Zhang Z. Human monocytes differentiate into tumor-associated macrophages upon SKOV3 cells coculture and/or lysophosphatidic acid stimulation. J Inflamm (Lond) 2022; 19:11. [PMID: 35842650 PMCID: PMC9288080 DOI: 10.1186/s12950-022-00307-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/06/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Serous ovarian carcinoma is the most common type of ovarian carcinoma. Tumor-associated macrophages (TAMs) promote ovarian cancer progression. Most macrophages are generated by monocyte differentiation. Lysophosphatidic acid (LPA) levels are high in blood, tissues and ascites of patients with ovarian cancer. This study investigated whether human monocytes can directly differentiate into TAMs in the serous ovarian carcinoma microenvironment. METHODS Human monocytes were isolated and purified from umbilical cord blood. A serous ovarian carcinoma-like microenvironment was generated by coculturing monocytes and SKOV3 cells in 0.4-μm-pore-size Transwell chambers. Additionally, the effect of LPA was assessed. The two cultured cell types and supernatants were evaluated. RESULTS The morphology and function of monocytes cocultured with SKOV3 cells and/or stimulated with LPA were significantly changed compared with those of non-stimulated monocytes. The CD14 + CD163 + and CD206 + phenotype indicated that stimulated cells were TAMs. The induced cells promoted SKOV3 cell proliferation and invasion, further proving that they were TAMs. The level of the cytokine interleukin-6R in the supernatant was significantly elevated in the treatment groups compared to the control monocyte group. Pathway enrichment analysis of ELISA results showed a strong influence of interleukin-6 family signaling, especially the JAK-STAT signaling pathway, further confirming the importance of IL-6R. CONCLUSION Monocytes can differentiate into TAMs under coculture with SKOV3 cells and/or LPA stimulation. The induced TAMs promote SKOV3 cell proliferation and invasion. The cytokine receptor IL-6sR and the JAK-STAT signaling pathway play an important role in the differentiation of monocytes into TAMs.
Collapse
Affiliation(s)
- Ying Feng
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, North Road of Workers Stadium, No. 8Chaoyang District, Beijing, 100020, China
- Department of Obstetrics and Gynecology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Meizhu Xiao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, North Road of Workers Stadium, No. 8Chaoyang District, Beijing, 100020, China
| | - Guangming Cao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, North Road of Workers Stadium, No. 8Chaoyang District, Beijing, 100020, China
| | - Hao Liu
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, North Road of Workers Stadium, No. 8Chaoyang District, Beijing, 100020, China
| | - Yanfang Li
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, North Road of Workers Stadium, No. 8Chaoyang District, Beijing, 100020, China
| | - Shuzhen Wang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, North Road of Workers Stadium, No. 8Chaoyang District, Beijing, 100020, China
| | - Stan Zijtveld
- Department of Obstetrics and Gynecology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Bert Delvoux
- Department of Obstetrics and Gynecology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Sofia Xanthoulea
- Department of Obstetrics and Gynecology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Andrea Romano
- Department of Obstetrics and Gynecology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Chongdong Liu
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, North Road of Workers Stadium, No. 8Chaoyang District, Beijing, 100020, China.
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, North Road of Workers Stadium, No. 8Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
59
|
Menghi F, Banda K, Kumar P, Straub R, Dobrolecki L, Rodriguez IV, Yost SE, Chandok H, Radke MR, Somlo G, Yuan Y, Lewis MT, Swisher EM, Liu ET. Genomic and epigenomic
BRCA
alterations predict adaptive resistance and response to platinum-based therapy in patients with triple-negative breast and ovarian carcinomas. Sci Transl Med 2022; 14:eabn1926. [PMID: 35857626 PMCID: PMC9585706 DOI: 10.1126/scitranslmed.abn1926] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Triple-negative breast cancer (TNBC) and ovarian carcinomas (OvCas) with
BRCA1
promoter methylation (
BRCA1
meth) respond more poorly to alkylating agents compared to those bearing mutations in
BRCA1
and
BRCA2
(
BRCA
mut). This is a conundrum given the biologically equivalent homologous recombination deficiency (HRD) induced by these genetic and epigenetic
BRCA
perturbations. We dissected this problem through detailed genomic analyses of TNBC and OvCa cohorts and experimentation with patient-derived xenografts and genetically engineered cell lines. We found that despite identical downstream genomic mutational signatures associated with
BRCA1
meth and
BRCA
mut states,
BRCA1
meth uniformly associates with poor outcomes. Exposure of
BRCA1
meth TNBCs to platinum chemotherapy, either as clinical treatment of a patient or as experimental in vivo exposure of preclinical patient derived xenografts, resulted in allelic loss of
BRCA1
methylation and increased
BRCA1
expression and platinum resistance. These data suggest that, unlike
BRCA
mut cancers, where
BRCA
loss is a genetically “fixed” deficiency state,
BRCA1
meth cancers are highly adaptive to genotoxin exposure and, through reversal of promoter methylation, recover
BRCA1
expression and become resistant to therapy. We further found a specific augmented immune transcriptional signal associated with enhanced response to platinum chemotherapy but only in patients with BRCA-proficient cancers. We showed how integrating both this cancer immune signature and the presence of
BRCA
mutations results in more accurate predictions of patient response when compared to either HRD status or
BRCA
status alone. This underscores the importance of defining
BRCA
heterogeneity in optimizing the predictive precision of assigning response probabilities in TNBC and OvCa.
Collapse
Affiliation(s)
- Francesca Menghi
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Kalyan Banda
- Division of Medical Oncology, UW Medical Center, Seattle, WA 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Pooja Kumar
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Robert Straub
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | | | - Isabel V. Rodriguez
- Department of Obstetrics and Gynecology, UW Medical Center, Seattle, WA 98195, USA
| | - Susan E. Yost
- Division of Medical Oncology and Therapeutic Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | | | - Marc R. Radke
- Department of Obstetrics and Gynecology, UW Medical Center, Seattle, WA 98195, USA
| | - George Somlo
- Division of Medical Oncology and Therapeutic Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Yuan Yuan
- Division of Medical Oncology and Therapeutic Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Michael T. Lewis
- Departments of Molecular and Cellular Biology and Radiology, Lester and Sue Smith Breast Center, Dan L Duncan Comprehensive Cancer Center, Houston, TX 77030, USA
| | - Elizabeth M. Swisher
- Department of Obstetrics and Gynecology, UW Medical Center, Seattle, WA 98195, USA
| | - Edison T. Liu
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| |
Collapse
|
60
|
Madeddu C, Donisi C, Liscia N, Lai E, Scartozzi M, Macciò A. EGFR-Mutated Non-Small Cell Lung Cancer and Resistance to Immunotherapy: Role of the Tumor Microenvironment. Int J Mol Sci 2022; 23:6489. [PMID: 35742933 PMCID: PMC9224267 DOI: 10.3390/ijms23126489] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is a leading cause of cancer-related deaths worldwide. About 10-30% of patients with non-small cell lung cancer (NSCLC) harbor mutations of the EGFR gene. The Tumor Microenvironment (TME) of patients with NSCLC harboring EGFR mutations displays peculiar characteristics and may modulate the antitumor immune response. EGFR activation increases PD-L1 expression in tumor cells, inducing T cell apoptosis and immune escape. EGFR-Tyrosine Kinase Inhibitors (TKIs) strengthen MHC class I and II antigen presentation in response to IFN-γ, boost CD8+ T-cells levels and DCs, eliminate FOXP3+ Tregs, inhibit macrophage polarization into the M2 phenotype, and decrease PD-L1 expression in cancer cells. Thus, targeted therapy blocks specific signaling pathways, whereas immunotherapy stimulates the immune system to attack tumor cells evading immune surveillance. A combination of TKIs and immunotherapy may have suboptimal synergistic effects. However, data are controversial because activated EGFR signaling allows NSCLC cells to use multiple strategies to create an immunosuppressive TME, including recruitment of Tumor-Associated Macrophages and Tregs and the production of inhibitory cytokines and metabolites. Therefore, these mechanisms should be characterized and targeted by a combined pharmacological approach that also concerns disease stage, cancer-related inflammation with related systemic symptoms, and the general status of the patients to overcome the single-drug resistance development.
Collapse
Affiliation(s)
- Clelia Madeddu
- Department of Medical Sciences and Public Health, Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, University of Cagliari, 09100 Cagliari, Italy; (C.M.); (N.L.); (E.L.); (M.S.)
| | - Clelia Donisi
- Department of Medical Sciences and Public Health, Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, University of Cagliari, 09100 Cagliari, Italy; (C.M.); (N.L.); (E.L.); (M.S.)
| | - Nicole Liscia
- Department of Medical Sciences and Public Health, Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, University of Cagliari, 09100 Cagliari, Italy; (C.M.); (N.L.); (E.L.); (M.S.)
| | - Eleonora Lai
- Department of Medical Sciences and Public Health, Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, University of Cagliari, 09100 Cagliari, Italy; (C.M.); (N.L.); (E.L.); (M.S.)
| | - Mario Scartozzi
- Department of Medical Sciences and Public Health, Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, University of Cagliari, 09100 Cagliari, Italy; (C.M.); (N.L.); (E.L.); (M.S.)
| | - Antonio Macciò
- Gynecologic Oncology Unit, ARNAS G. Brotzu, Department of Surgical Sciences, University of Cagliari, 09100 Cagliari, Italy;
| |
Collapse
|
61
|
Cao X, Lai SWT, Chen S, Wang S, Feng M. Targeting tumor-associated macrophages for cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:61-108. [PMID: 35636930 DOI: 10.1016/bs.ircmb.2022.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Tumor-associated macrophages (TAMs) are one of the most abundant immune components in the tumor microenvironment and play a plethora of roles in regulating tumorigenesis. Therefore, the therapeutic targeting of TAMs has emerged as a new paradigm for immunotherapy of cancer. Herein, the review summarizes the origin, polarization, and function of TAMs in the progression of malignant diseases. The understanding of such knowledge leads to several distinct therapeutic strategies to manipulate TAMs to battle cancer, which include those to reduce TAM abundance, such as depleting TAMs or inhibiting their recruitment and differentiation, and those to harness or boost the anti-tumor activities of TAMs such as blocking phagocytosis checkpoints, inducing antibody-dependent cellular phagocytosis, and reprogramming TAM polarization. In addition, modulation of TAMs may reshape the tumor microenvironment and therefore synergize with other cancer therapeutics. Therefore, the rational combination of TAM-targeting therapeutics with conventional therapies including radiotherapy, chemotherapy, and other immunotherapies is also reviewed. Overall, targeting TAMs presents itself as a promising strategy to add to the growing repertoire of treatment approaches in the fight against cancer, and it is hopeful that these approaches currently being pioneered will serve to vastly improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| | - Seigmund W T Lai
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Siqi Chen
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Sadira Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| |
Collapse
|
62
|
Spheroid Formation and Peritoneal Metastasis in Ovarian Cancer: The Role of Stromal and Immune Components. Int J Mol Sci 2022; 23:ijms23116215. [PMID: 35682890 PMCID: PMC9181487 DOI: 10.3390/ijms23116215] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common gynecological cancers, with the worst prognosis and the highest mortality rate. Peritoneal dissemination (or carcinomatosis) accompanied by ascites formation is the most unfavorable factor in the progression and recurrence of OC. Tumor cells in ascites are present as either separate cells or, more often, as cell aggregates, i.e., spheroids which promote implantation on the surface of nearby organs and, at later stages, metastases to distant organs. Malignant ascites comprises a unique tumor microenvironment; this fact may be of relevance in the search for new prognostic and predictive factors that would make it possible to personalize the treatment of patients with OC. However, the precise mechanisms of spheroid formation and carcinomatosis are still under investigation. Here, we summarize data on ascites composition as well as the activity of fibroblasts and macrophages, the key stromal and immune components, in OC ascites. We describe current knowledge about the role of fibroblasts and macrophages in tumor spheroid formation, and discuss the specific functions of fibroblasts, macrophages and T cells in tumor peritoneal dissemination and implantation.
Collapse
|
63
|
Su S, Lei A, Wang X, Lu H, Wang S, Yang Y, Li N, Zhang Y, Zhang J. Induced CAR-Macrophages as a Novel Therapeutic Cell Type for Cancer Immune Cell Therapies. Cells 2022; 11:1652. [PMID: 35626689 PMCID: PMC9139529 DOI: 10.3390/cells11101652] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/09/2022] [Accepted: 04/22/2022] [Indexed: 01/27/2023] Open
Abstract
The Chimeric antigen receptor (CAR)-T cell therapy has made inroads in treating hematological malignancies. Nonetheless, there are still multiple hurdles in CAR-T cell therapy for solid tumors. Primary CAR-expressing macrophage cells (CAR-Ms) and induced pluripotent stem cells (iPSCs)-derived CAR-expressing macrophage cells (CAR-iMacs) have emerged as attractive alternatives in our quest for an efficient and inexpensive approach for tumor immune cell therapy. In this review, we list the current state of development of human CAR-macrophages and provide an overview of the crucial functions of human CAR-macrophages in the field of tumor immune cell therapy.
Collapse
Affiliation(s)
- Siyu Su
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China;
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; (A.L.); (X.W.); (H.L.)
| | - Anhua Lei
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; (A.L.); (X.W.); (H.L.)
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou 310058, China
| | - Xudong Wang
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; (A.L.); (X.W.); (H.L.)
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou 310058, China
| | - Hengxing Lu
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; (A.L.); (X.W.); (H.L.)
| | - Shuhang Wang
- National Cancer Center/National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Beijing 100021, China; (S.W.); (N.L.)
| | - Yuqi Yang
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People’s Hospital, No. 83 Zhongshan Road, Guiyang 550000, China;
| | - Ning Li
- National Cancer Center/National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Beijing 100021, China; (S.W.); (N.L.)
| | - Yi Zhang
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China;
| | - Jin Zhang
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; (A.L.); (X.W.); (H.L.)
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
64
|
Zhang J, Li S, Liu F, Yang K. Role of CD68 in tumor immunity and prognosis prediction in pan-cancer. Sci Rep 2022; 12:7844. [PMID: 35550532 PMCID: PMC9098459 DOI: 10.1038/s41598-022-11503-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 04/11/2022] [Indexed: 12/24/2022] Open
Abstract
CD68 plays a critical role in promoting phagocytosis; however, the function of CD68 in tumor immunity and prognosis remains unknown. We analyzed CD68 expression among 33 tumor and normal tissues from The Cancer Genome Atlas and Genotype-Tissue Expression datasets. The relationship between CD68 expression and cancer prognosis, immune infiltration, checkpoint markers, and drug response was explored. Upregulated CD68 levels were observed in various cancer types, which were verified through tumor tissue chips using immunohistochemistry. High levels of CD68 in tumor samples correlated with an adverse prognosis in glioblastoma, kidney renal clear cell carcinoma, lower-grade glioma, liver hepatocellular carcinoma, lung squamous cell carcinoma, thyroid carcinoma, and thymoma and a favorable prognosis in kidney chromophobe. The top three negatively enriched Kyoto Encyclopedia of Genes and Genomes terms in the high CD68 subgroup were chemokine signaling pathway, cytokine-cytokine receptor interaction, and cell adhesion molecule cams. The top negatively enriched HALLMARK terms included complement, allograft rejection, and inflammatory response. A series of targeted drugs and small-molecule drugs with promising therapeutic effects were predicted. The clinical prognosis and immune infiltration of high expression levels of CD68 differ across tumor types. Inhibiting CD68-dependent signaling could be a promising therapeutic strategy for immunotherapy in many tumor types.
Collapse
Affiliation(s)
- Jingwei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Shuwang Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Kui Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.
| |
Collapse
|
65
|
Glass EB, Hoover AA, Bullock KK, Madden MZ, Reinfeld BI, Harris W, Parker D, Hufnagel DH, Crispens MA, Khabele D, Rathmell WK, Rathmell JC, Wilson AJ, Giorgio TD, Yull FE. Stimulating TAM-mediated anti-tumor immunity with mannose-decorated nanoparticles in ovarian cancer. BMC Cancer 2022; 22:497. [PMID: 35513776 PMCID: PMC9074180 DOI: 10.1186/s12885-022-09612-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/21/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Current cancer immunotherapies have made tremendous impacts but generally lack high response rates, especially in ovarian cancer. New therapies are needed to provide increased benefits. One understudied approach is to target the large population of immunosuppressive tumor-associated macrophages (TAMs). Using inducible transgenic mice, we recently reported that upregulating nuclear factor-kappaB (NF-κB) signaling in TAMs promotes the M1, anti-tumor phenotype and limits ovarian cancer progression. We also developed a mannose-decorated polymeric nanoparticle system (MnNPs) to preferentially deliver siRNA payloads to M2, pro-tumor macrophages in vitro. In this study, we tested a translational strategy to repolarize ovarian TAMs via MnNPs loaded with siRNA targeting the inhibitor of NF-κB alpha (IκBα) using mouse models of ovarian cancer. METHODS We evaluated treatment with MnNPs loaded with IκBα siRNA (IκBα-MnNPs) or scrambled siRNA in syngeneic ovarian cancer models. ID8 tumors in C57Bl/6 mice were used to evaluate consecutive-day treatment of late-stage disease while TBR5 tumors in FVB mice were used to evaluate repetitive treatments in a faster-developing disease model. MnNPs were evaluated for biodistribution and therapeutic efficacy in both models. RESULTS Stimulation of NF-κB activity and repolarization to an M1 phenotype via IκBα-MnNP treatment was confirmed using cultured luciferase-reporter macrophages. Delivery of MnNPs with fluorescent payloads (Cy5-MnNPs) to macrophages in the solid tumors and ascites was confirmed in both tumor models. A three consecutive-day treatment of IκBα-MnNPs in the ID8 model validated a shift towards M1 macrophage polarization in vivo. A clear therapeutic effect was observed with biweekly treatments over 2-3 weeks in the TBR5 model where significantly reduced tumor burden was accompanied by changes in immune cell composition, indicative of reduced immunosuppressive tumor microenvironment. No evidence of toxicity associated with MnNP treatment was observed in either model. CONCLUSIONS In mouse models of ovarian cancer, MnNPs were preferentially associated with macrophages in ascites fluid and solid tumors. Evidence of macrophage repolarization, increased inflammatory cues, and reduced tumor burden in IκBα-MnNP-treated mice indicate beneficial outcomes in models of established disease. We have provided evidence of a targeted, TAM-directed approach to increase anti-tumor immunity in ovarian cancer with strong translational potential for future clinical studies.
Collapse
Affiliation(s)
- Evan B Glass
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Alyssa A Hoover
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Kennady K Bullock
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Matthew Z Madden
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Bradley I Reinfeld
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Whitney Harris
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Dominique Parker
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | | | - Marta A Crispens
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dineo Khabele
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - W Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew J Wilson
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Todd D Giorgio
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Fiona E Yull
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
66
|
Epigenomic Profiling of Epithelial Ovarian Cancer Stem-Cell Differentiation Reveals GPD1 Associated Immune Suppressive Microenvironment and Poor Prognosis. Int J Mol Sci 2022; 23:ijms23095120. [PMID: 35563509 PMCID: PMC9101898 DOI: 10.3390/ijms23095120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/03/2022] [Accepted: 05/03/2022] [Indexed: 11/21/2022] Open
Abstract
Intraperitoneal metastasis is a challenging clinical scenario in epithelial ovarian cancer (EOC). As they are distinct from hematogenous metastasizing tumors, epithelial ovarian cancer cells primarily disseminate within the peritoneal cavity to form superficially invasive carcinomas. Unfavorable pharmacokinetics for peritoneal tumors and gut toxicity collectively lead to a narrow therapeutic window and therefore limit the opportunities for a favorable clinical outcome. New insights into tumor metastasis in the peritoneal microenvironment are keenly awaited to develop new therapeutic strategies. Epithelial ovarian cancer stem cell (OCSC) seeding is considered to be a critical component of the peritoneal spread. Using a unique and stepwise process of the OCSC differentiation model may provide insight into the intraperitoneal metastasis. The transcriptome and epigenome of OCSC differentiation were characterized by expression array and MethylCap-Seq. The TCGA, AOCS, and KM-Plotter databases were used to evaluate the association between survival outcomes and the methylation/expression levels of candidate genes in the EOC datasets. The STRING database was used to investigate the protein–protein interaction (PPI) for candidates and their associated genes. The infiltration level of immune cells in EOC patients and the association between clinical outcome and OCSCs differentiation genes were estimated using the TIDE and TIME2.0 algorithms. We established an EOC differentiation model using OCSCs. After an integrated transcriptomics and methylomics analysis of OCSCs differentiation, we revealed that the genes associated with earlier OCSC differentiation were better able to reflect the patient’s outcome. The OCSC differentiation genes were involved in regulating metabolism shift and the suppressive immune microenvironment. High GPD1 expression with high pro-tumorigenic immune cells (M2 macrophage, and cancer associated fibroblast) had worst survival. Moreover, we developed a methylation signature, constituted by GNPDA1, GPD1, GRASP, HOXC11, and MSLN, that may be useful for prognostic prediction in EOC. Our results revealed a novel role of epigenetic plasticity OCSC differentiation and suggested metabolic and immune intervention as a new therapeutic strategy.
Collapse
|
67
|
Sidiropoulos DN, Rafie CI, Jang JK, Castanon S, Baugh AG, Gonzalez E, Christmas BJ, Narumi VH, Davis-Marcisak EF, Sharma G, Bigelow E, Vaghasia A, Gupta A, Skaist A, Considine M, Wheelan SJ, Ganesan SK, Yu M, Yegnasubramanian S, Stearns V, Connolly RM, Gaykalova DA, Kagohara LT, Jaffee EM, Fertig EJ, Roussos Torres ET. Entinostat Decreases Immune Suppression to Promote Antitumor Responses in a HER2+ Breast Tumor Microenvironment. Cancer Immunol Res 2022; 10:656-669. [PMID: 35201318 PMCID: PMC9064912 DOI: 10.1158/2326-6066.cir-21-0170] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 10/19/2021] [Accepted: 02/18/2022] [Indexed: 11/16/2022]
Abstract
Therapeutic combinations to alter immunosuppressive, solid tumor microenvironments (TME), such as in breast cancer, are essential to improve responses to immune checkpoint inhibitors (ICI). Entinostat, an oral histone deacetylase inhibitor, has been shown to improve responses to ICIs in various tumor models with immunosuppressive TMEs. The precise and comprehensive alterations to the TME induced by entinostat remain unknown. Here, we employed single-cell RNA sequencing on HER2-overexpressing breast tumors from mice treated with entinostat and ICIs to fully characterize changes across multiple cell types within the TME. This analysis demonstrates that treatment with entinostat induced a shift from a protumor to an antitumor TME signature, characterized predominantly by changes in myeloid cells. We confirmed myeloid-derived suppressor cells (MDSC) within entinostat-treated tumors associated with a less suppressive granulocytic (G)-MDSC phenotype and exhibited altered suppressive signaling that involved the NFκB and STAT3 pathways. In addition to MDSCs, tumor-associated macrophages were epigenetically reprogrammed from a protumor M2-like phenotype toward an antitumor M1-like phenotype, which may be contributing to a more sensitized TME. Overall, our in-depth analysis suggests that entinostat-induced changes on multiple myeloid cell types reduce immunosuppression and increase antitumor responses, which, in turn, improve sensitivity to ICIs. Sensitization of the TME by entinostat could ultimately broaden the population of patients with breast cancer who could benefit from ICIs.
Collapse
Affiliation(s)
- Dimitrios N Sidiropoulos
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | | | - Julie K Jang
- Division of Medical Oncology, Department of Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Sofi Castanon
- Division of Medical Oncology, Department of Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Aaron G Baugh
- Division of Medical Oncology, Department of Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Edgar Gonzalez
- Division of Medical Oncology, Department of Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Brian J Christmas
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Valerie H Narumi
- Division of Medical Oncology, Department of Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Emily F Davis-Marcisak
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gaurav Sharma
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Emma Bigelow
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Ajay Vaghasia
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Anuj Gupta
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Alyza Skaist
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Michael Considine
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Sarah J Wheelan
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Sathish Kumar Ganesan
- Department of Stem Cell Biology and Regenerative Medicine, and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Srinivasan Yegnasubramanian
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Vered Stearns
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Roisin M Connolly
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Cancer Research at UCC, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Daria A Gaykalova
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland Medical Center, Baltimore, Maryland
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, Maryland
- Institute for Genome Sciences, University of Maryland Medical Center, Baltimore, Maryland
| | - Luciane T Kagohara
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Elana J Fertig
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, Maryland
| | - Evanthia T Roussos Torres
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Division of Medical Oncology, Department of Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
68
|
Shah D, Challagundla N, Dave V, Patidar A, Saha B, Nivsarkar M, Trivedi VB, Agrawal-Rajput R. Berberine mediates tumor cell death by skewing tumor-associated immunosuppressive macrophages to inflammatory macrophages. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:153904. [PMID: 35231825 DOI: 10.1016/j.phymed.2021.153904] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 12/11/2021] [Accepted: 12/19/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Berberine is a plant-derived alkaloid with potent anti-cancer activities. Berberine may redirect the tumor-promoting immunosuppressive M2 macrophages, to tumoricidal activated M1 macrophages. But such an anti-tumor function remains to be demonstrated. HYPOTHESIS Polarization of macrophages to an immunosuppressive phenotype within the tumor microenvironment promotes tumor growth and contributes to resistance to chemotherapy. We examined if berberine would target macrophage polarization to reinstate anti-tumor immune response. STUDY DESIGN Using a B16F10 mouse melanoma model, we assessed berberine-induced re-polarization of immunosuppressive M2 macrophages to anti-tumor M1 macrophages and subsequent T-cell activation within the immunosuppressive tumor microenvironment. METHODS The B16F10 culture supernatant along with tumor antigen was used as tumor mimicking conditioned medium (CM). The bone marrow-derived macrophages were cultured in CM for 5 days. The CM-induced skewing of macrophages to M2-like phenotype was confirmed by flow cytometry and ELISA. The T-cells were co-cultured with macrophages to decipher the effect of berberine on T-cell differentiation. In vivo efficacy of berberine was analyzed using melanoma model of solid tumor. RESULTS Berberine inhibited rIL-6-induced STAT-3 phosphorylation and IL-10 release from B16F10 cells. It enhanced tumor antigen-induced IL-1β, IL-12 and TNFα, but suppressed IL-6 and TGF-β release. Berberine significantly prevented the tumor antigen-mediated IL-10-enhanced IL-6 and TGF-β expression. The CM skewed the bone marrow-derived macrophages to CD206-high but MHC-II-low M2-like tumor-associated macrophages. Berberine partially prevented the generation of these macrophages and was associated with reduced C/EBPβ and Egr2 mRNA expression and lowered IL-10 and TGF-β production. Berberine significantly reduced Arginase-1 expression in CM-treated M1 and M2-like macrophages. Berberine increased MHC-II and CD40 expression on the macrophages augmenting the CTL activity and the number of IFNγ-producing CD4+ T-cells. Berberine significantly lowered tumor volume, weight and enhanced the frequency of M1-like macrophages in mice. CONCLUSION These data indicate that berberine interferes with pro-tumor macrophage polarization and IL-10 and TGF-β release but restores Tcell anti-tumor cytotoxicity in the tumor microenvironment.
Collapse
Affiliation(s)
- Dhruvi Shah
- Immunology Lab, Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat 382 426, India
| | - Naveen Challagundla
- Immunology Lab, Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat 382 426, India
| | - Vaidehi Dave
- Immunology Lab, Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat 382 426, India
| | - Ashok Patidar
- National Centre for Cell Science (NCCS), Pune 411 007, India
| | - Bhaskar Saha
- National Centre for Cell Science (NCCS), Pune 411 007, India
| | - Manish Nivsarkar
- Department of Pharmacology and Toxicology, B.V. Patel PERD Centre, Ahmedabad, Gujarat 380 054, India
| | - Varsha B Trivedi
- Department of Pathology, Laboratory Medicine, Transfusion Services and Immuno haematology, IKDRC-ITS, Ahmedabad 380 016, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat 382 426, India.
| |
Collapse
|
69
|
Musick M, Yu X. Manipulation of the tumor immuno-microenvironment via TAM-targeted expression of transcription factors. Immunol Res 2022; 70:432-440. [PMID: 35486115 DOI: 10.1007/s12026-022-09277-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/01/2022] [Indexed: 11/28/2022]
Abstract
An immunosuppressive tumor microenvironment (TME) leads to cancer growth, metastasis, and therapeutic resistance. Immunomodulatory immunotherapy aims to skew the immunosuppressive TME back to an immune active state. Tumor-associated macrophages (TAMs) are a critical component of the TME that are actively involved in tumor-specific inflammation and immunosuppression. TAMs exhibit a diverse range of phenotypes and functions, from pro-tumor to anti-tumor. The plasticity of TAMs makes them a promising target for immunotherapy, and TAM-targeted therapies via different strategies have shown great potential. This review discusses current TAM-specific delivery targets and genes of interest for TAM-reprogramming. As phagocytic cells, TAMs have several receptors that have been used to increase TAM-targeted in vivo delivery. Furthermore, a promising approach for reprogramming TAMs is to activate or suppress specific transcription factors in the signal transducers and activators of transcription (STAT) and interferon regulatory factor (IRF) families. Altering TAM transcription factor expression results in a potent shift in cytokine expression and overall TAM function potentially tipping the balance from an immunosuppressive to an immune active TME.
Collapse
Affiliation(s)
- Maggie Musick
- Department of Biological Sciences, Clemson University, 132 Long Hall, SC, 29631, Clemson, USA.
| | - Xianzhong Yu
- Department of Biological Sciences, Clemson University, 132 Long Hall, SC, 29631, Clemson, USA
| |
Collapse
|
70
|
Kim T, Johnston J, Castillo-Lluva S, Cimas FJ, Hamby S, Gonzalez-Moreno S, Villarejo-Campos P, Goodall AH, Velasco G, Ocana A, Muthana M, Kiss-Toth E. TRIB1 regulates tumor growth via controlling tumor-associated macrophage phenotypes and is associated with breast cancer survival and treatment response. Theranostics 2022; 12:3584-3600. [PMID: 35664073 PMCID: PMC9131267 DOI: 10.7150/thno.72192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/05/2022] [Indexed: 11/05/2022] Open
Abstract
Molecular mechanisms that regulate tumor-associated macrophage (TAM) phenotype and function are incompletely understood. The pseudokinase TRIB1 has been reported as a regulator of macrophage phenotypes, both in mouse and human systems. Methods: Bioinformatic analysis was used to investigate the link between TRIB1 expression in breast cancer and therapeutic response to chemotherapy. In vivo models of breast cancer included immune-competent mice to characterize the consequences of altered (reduced or elevated) myeloid Trib1 expression on tumor growth and composition of stromal immune cell populations. Results: TRIB1 was highly expressed by TAMs in breast cancer and high TRIB1 expression correlated with response to chemotherapy and patient survival. Both overexpression and knockout of myeloid Trib1 promote mouse breast tumor growth, albeit through different molecular mechanisms. Myeloid Trib1 deficiency led to an early acceleration of tumor growth, paired with a selective reduction in perivascular macrophage numbers in vivo and enhanced oncogenic cytokine expression in vitro. In contrast, elevated levels of Trib1 in myeloid cells led to an increased late-stage mammary tumor volume, coupled with a reduction of NOS2 expressing macrophages and an overall reduction of macrophages in hypoxic tumor regions. In addition, we show that myeloid Trib1 is a previously unknown, negative regulator of the anti-tumor cytokine IL-15, and that increased myeloid Trib1 expression leads to reduced IL-15 levels in mammary tumors, with a consequent reduction in the number of T-cells that are key to anti-tumor immune responses. Conclusions: Together, these results define a key role for TRIB1 in chemotherapy responses for human breast cancer and provide a mechanistic understanding for the importance of the control of myeloid TRIB1 expression in the development of this disease.
Collapse
Affiliation(s)
- Taewoo Kim
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield Medical School, Sheffield, S10 2RX, UK
| | - Jessica Johnston
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield Medical School, Sheffield, S10 2RX, UK
| | - Sonia Castillo-Lluva
- Department of Biochemistry and Molecular Biology, Complutense University and Instituto de Investigación Sanitaria Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Francisco J Cimas
- Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria Clínico San Carlos (IdISSC), Madrid and Universidad de Castilla La Mancha (UCLM), Albacete, Spain
| | - Stephen Hamby
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester and Leicester NIHR Biomedical Research Centre, Leicester, UK
| | | | | | - Alison H Goodall
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester and Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology, Complutense University and Instituto de Investigación Sanitaria Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Alberto Ocana
- Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria Clínico San Carlos (IdISSC), Madrid and Universidad de Castilla La Mancha (UCLM), Albacete, Spain
| | - Munitta Muthana
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, S10 2RX, UK
| | - Endre Kiss-Toth
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield Medical School, Sheffield, S10 2RX, UK
- Biological Research Centre of the Hungarian Academy of Sciences, Temesvari krt. 62, Szeged, 6726, Hungary
| |
Collapse
|
71
|
Dunn ZS, Li YR, Yu Y, Lee D, Gibbons A, Kim JJ, Zhou TY, Li M, Nguyen M, Cen X, Zhou Y, Wang P, Yang L. Minimally Invasive Preclinical Monitoring of the Peritoneal Cavity Tumor Microenvironment. Cancers (Basel) 2022; 14:1775. [PMID: 35406547 PMCID: PMC8997523 DOI: 10.3390/cancers14071775] [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: 03/07/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 11/24/2022] Open
Abstract
Intraperitoneal (i.p.) experimental models in mice can recapitulate the process of i.p. dissemination in abdominal cancers and may help uncover critical information about future successful clinical treatments. i.p. cellular composition is studied in preclinical models addressing a wide spectrum of other pathophysiological states such as liver cirrhosis, infectious disease, autoimmunity, and aging. The peritoneal cavity is a multifaceted microenvironment that contains various immune cell populations, including T, B, NK, and various myeloid cells, such as macrophages. Analysis of the peritoneal cavity is often obtained by euthanizing mice and performing terminal peritoneal lavage. This procedure inhibits continuous monitoring of the peritoneal cavity in a single mouse and necessitates the usage of more mice to assess the cavity at multiple timepoints, increasing the cost, time, and variability of i.p. studies. Here, we present a simple, novel method termed in vivo intraperitoneal lavage (IVIPL) for the minimally invasive monitoring of cells in the peritoneal cavity of mice. In this proof-of-concept, IVIPL provided real-time insights into the i.p. tumor microenvironment for the development and study of ovarian cancer therapies. Specifically, we studied CAR-T cell therapy in a human high-grade serous ovarian cancer (HGSOC) xenograft mouse model, and we studied the immune composition of the i.p. tumor microenvironment (TME) in a mouse HGSOC syngeneic model.
Collapse
Affiliation(s)
- Zachary Spencer Dunn
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA; (Z.S.D.); (P.W.)
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Yanqi Yu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Derek Lee
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Alicia Gibbons
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - James Joon Kim
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Tian Yang Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Mulin Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Mya Nguyen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Xinjian Cen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Yang Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA; (Z.S.D.); (P.W.)
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
72
|
Shen X, Gu X, Ma R, Li X, Wang J. Identification of the Immune Signatures for Ovarian Cancer Based on the Tumor Immune Microenvironment Genes. Front Cell Dev Biol 2022; 10:772701. [PMID: 35372348 PMCID: PMC8974491 DOI: 10.3389/fcell.2022.772701] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 03/02/2022] [Indexed: 11/21/2022] Open
Abstract
Ovarian cancer (OV) is a deadly gynecological cancer. The tumor immune microenvironment (TIME) plays a pivotal role in OV development. However, the TIME of OV is not fully known. Therefore, we aimed to provide a comprehensive network of the TIME in OV. Gene expression data and clinical information from OV patients were obtained from the Cancer Genome Atlas Program (TCGA) database. Non-negative Matrix Factorization, NMFConsensus, and nearest template prediction algorithms were used to perform molecular clustering. The biological functions of differentially expressed genes (DEGs) were identified using Metascape, gene set enrichment analysis (GSEA), gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The copy number variations (CNVs), single nucleotide polymorphisms (SNPs) and tumor mutation burden were analyzed using Gistic 2.0, R package maftools, and TCGA mutations, respectively. Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data and CIBERSORT were utilized to elucidate the TIME. Moreover, external data from the International Cancer Genome Consortium (ICGC) and ArrayExpress databases were used to validate the signature. All 361 samples from the TCGA OV dataset were classified into Immune Class and non-Immune Class with immune signatures. By comparing the two classes, we identified 740 DEGs that accumulated in immune-related, cancer-related, inflammation-related biological functions and pathways. There were significant differences in the CNVs between the Immune and non-Immune Classes. The Immune Class was further divided into immune-activated and immune-suppressed subtypes. There was no significant difference in the top 20 genes in somatic SNPs among the three groups. In addition, the immune-activated subtype had significantly increased proportions of CD4 memory resting T cells, T cells, M1 macrophages, and M2 macrophages than the other two groups. The qRT-PCR results indicated that the mRNA expression levels of RYR2, FAT3, MDN1 and RYR1 were significantly down-regulated in OV compared with normal tissues. Moreover, the signatures of the TIME were validated using ICGC cohort and the ArrayExpress cohort. Our study clustered the OV patients into an immune-activated subtype, immune-suppressed subtype, and non-Immune Class and provided potential clues for further research on the molecular mechanisms and immunotherapy strategies of OV.
Collapse
Affiliation(s)
- Xiaoyan Shen
- Department of Gynecology, Peking University People’s Hospital, Beijing, China
| | - Xiao Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ruiqiong Ma
- Department of Gynecology, Peking University People’s Hospital, Beijing, China
| | - Xiaoping Li
- Department of Gynecology, Peking University People’s Hospital, Beijing, China
| | - Jianliu Wang
- Department of Gynecology, Peking University People’s Hospital, Beijing, China
- *Correspondence: Jianliu Wang,
| |
Collapse
|
73
|
Zhang J, Wang Z, Zhang H, Dai Z, Liang X, Li S, Zhang X, Liu F, Liu Z, Yang K, Cheng Q. Functions of RNF Family in the Tumor Microenvironment and Drugs Prediction in Grade II/III Gliomas. Front Cell Dev Biol 2022; 9:754873. [PMID: 35223862 PMCID: PMC8864229 DOI: 10.3389/fcell.2021.754873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence has demonstrated that RING finger (RNF) proteins played a vital role in cellular and physiological processes and various diseases. However, the function of RNF proteins in low-grade glioma (LGG) remains unknown. In this study, 138 RNF family members revealed their role in LGG. The TCGA database was used as the training cohort; two CGGA databases and GSE108474 were selected as external validation cohorts. Patients were grouped into cluster 1 and cluster 2, both in the training and validation cohorts, using consensus clustering analysis. The prognosis of patients in cluster 1 is significantly better than that in cluster 2. Meanwhile, biofunction prediction was further introduced to explore the potential mechanisms that led to differences in survival outcomes. Patients in Cluster 2 showed more complicated immunocytes infiltration and highly immunosuppressive features than cluster 1. Enrichment pathways such as negative regulation of mast cell activation, DNA replication, mismatch repair, Th17 cell differentiation, antigen processing and presentation, dendritic cell antigen processing and presentation, dendritic cell differentiation were also enriched in cluster 2 patients. For the last, the main contributors were distinguished by employing a machine learning algorithm. A lot of targeted and small molecule drugs that are sensitive to patients in cluster 2 were predicted. Importantly, we discovered TRIM8, DTX2, and TRAF5 as the most vital contributors from the RNF family, which were related to immune infiltration in LGG tumor immune landscape. In this study, we demonstrated the predicted role of RNF proteins in LGG. In addition, we found out three markers among RNF proteins that are closely related to the immune aspects of LGG, which might serve as novel therapeutic targets for immunotherapy in the future.
Collapse
Affiliation(s)
- Jingwei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuwang Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xun Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Clinical Diagnosis and Therapy Center for Glioma of Xiangya Hospital, Central South University, Changsha, China
| | - Kui Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Clinical Diagnosis and Therapy Center for Glioma of Xiangya Hospital, Central South University, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
74
|
Targeting CCR2+ macrophages with BET inhibitor overcomes adaptive resistance to anti-VEGF therapy in ovarian cancer. J Cancer Res Clin Oncol 2022; 148:803-821. [PMID: 35094142 PMCID: PMC8930900 DOI: 10.1007/s00432-021-03885-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/11/2021] [Indexed: 12/14/2022]
Abstract
Purpose Tumor-associated macrophages (TAMs) are known to contribute to adaptive resistance to anti-vascular endothelial growth factor (VEGF) antibody (AVA) therapy in ovarian cancer. BET (bromodomain and extra-terminal domain) inhibitors (BETi) may have unique roles in targeting TAMs. Our objective was to examine the effects of BETi on TAMs, especially in the context of enhancing the efficacy of AVA therapy. Methods We conducted a series of in vitro (MTT assay, apoptosis, flow cytometry, and RNA sequencing) and in vivo (xenograft ovarian cancer model) experiments to determine the biological effects of BETi combined with AVA in ovarian cancer. For statistical analysis, a two-tailed Student’s t test (equal variance) or ANOVA was used for multiple groups’ comparison, and p < 0.05 was considered significant. Results BETi resulted in a dose-dependent decrease in cell viability and induced apoptosis (p < 0.01) in ovarian cancer cells (SKOV3ip1, OVCAR5, and OVCAR8). Treatment with BETi significantly increased apoptosis in THP-1 monocytes and macrophages (PMA-differentiated THP-1; p < 0.01). Furthermore, BETi selectively induced greater apoptosis in M2-like macrophages (PMA and IL-4, IL-13-differentiated THP-1) (31.3%-36.1%) than in M1-like macrophages (PMA and LPS-differentiated THP-1) (12.4%-18.5%) (p < 0.01). Flow cytometry revealed that the percentage of M1-like macrophages (CD68+/CD80+) was significantly increased after treatment with low-dose BETi (ABBV-075 0.1 µM; p < 0.05), whereas the percentage of CD68+/CCR2+ macrophages was significantly decreased (p < 0.001); these findings suggest that BETi may selectively inhibit the survival of CCR2+ macrophages and re-polarize the macrophages into an M1-like phenotype. RNA-seq analysis revealed that BETi selectively targeted macrophage infiltration-related cytokines/chemokines in ovarian cancer (adjusted p < 0.05 and Log2 fold change ≥ 1.5). Finally, using in vivo ovarian cancer models, compared with control or monotherapy, the combination of BETi (ABBV-075) and bevacizumab resulted in greater inhibition of tumor growth and macrophage infiltration (p < 0.05) and longer survival of tumor-bearing mice (p < 0.001). Conclusions Our findings indicate a previously unrecognized role for BETi in selectively targeting CCR2+ TAMs and enhancing the efficacy of AVA therapy in ovarian cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03885-z.
Collapse
|
75
|
Kenry, Eschle BK, Andreiuk B, Gokhale PC, Mitragotri S. Differential Macrophage Responses to Gold Nanostars and Their Implication for Cancer Immunotherapy. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kenry
- Harvard John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Department of Imaging Dana‐Farber Cancer Institute and Harvard Medical School Boston MA 02215 USA
| | - Benjamin K. Eschle
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science Dana‐Farber Cancer Institute Boston MA 02215 USA
| | - Bohdan Andreiuk
- Department of Imaging Dana‐Farber Cancer Institute and Harvard Medical School Boston MA 02215 USA
- Department of Cancer Immunology and Virology Dana‐Farber Cancer Institute and Harvard Medical School Boston MA 02215 USA
| | - Prafulla C. Gokhale
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science Dana‐Farber Cancer Institute Boston MA 02215 USA
| | - Samir Mitragotri
- Harvard John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| |
Collapse
|
76
|
Szulc-Kielbik I, Kielbik M. Tumor-Associated Macrophages: Reasons to Be Cheerful, Reasons to Be Fearful. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 113:107-140. [PMID: 35165862 DOI: 10.1007/978-3-030-91311-3_4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor microenvironment (TME) is a complex and constantly evolving entity that consists not only of cancer cells, but also of resident host cells and immune-infiltrating cells, among which macrophages are significant components, due to their diversity of functions through which they can influence the immune response against tumor cells. Macrophages present in tumor environment are termed as tumor-associated macrophages (TAMs). They are strongly plastic cells, and depending on the TME stimuli (i.e., cytokines, chemokines), TAMs polarize to antitumoral (M1-like TAMs) or protumoral (M2-like TAMs) phenotype. Both types of TAMs differ in the surface receptors' expression, activation of intracellular signaling pathways, and ability of production and various metabolites release. At the early stage of tumor formation, TAMs are M1-like phenotype, and they are able to eliminate tumor cells, i.e., by reactive oxygen species formation or by presentation of cancer antigens to other effector immune cells. However, during tumor progression, TAMs M2-like phenotype is dominating. They mainly contribute to angiogenesis, stromal remodeling, enhancement of tumor cells migration and invasion, and immunosuppression. This wide variety of TAMs' functions makes them an excellent subject for use in developing antitumor therapies which mainly is based on three strategies: TAMs' elimination, reprograming, or recruitment inhibition.
Collapse
Affiliation(s)
| | - Michal Kielbik
- Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland.
| |
Collapse
|
77
|
Miyamoto T, Murakami R, Hamanishi J, Tanigaki K, Hosoe Y, Mise N, Takamatsu S, Mise Y, Ukita M, Taki M, Yamanoi K, Horikawa N, Abiko K, Yamaguchi K, Baba T, Matsumura N, Mandai M. B7-H3 Suppresses Antitumor Immunity via the CCL2-CCR2-M2 Macrophage Axis and Contributes to Ovarian Cancer Progression. Cancer Immunol Res 2022; 10:56-69. [PMID: 34799346 PMCID: PMC9414298 DOI: 10.1158/2326-6066.cir-21-0407] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/03/2021] [Accepted: 11/18/2021] [Indexed: 01/07/2023]
Abstract
New approaches beyond PD-1/PD-L1 inhibition are required to target the immunologically diverse tumor microenvironment (TME) in high-grade serous ovarian cancer (HGSOC). In this study, we explored the immunosuppressive effect of B7-H3 (CD276) via the CCL2-CCR2-M2 macrophage axis and its potential as a therapeutic target. Transcriptome analysis revealed that B7-H3 is highly expressed in PD-L1-low, nonimmunoreactive HGSOC tumors, and its expression negatively correlated with an IFNγ signature, which reflects the tumor immune reactivity. In syngeneic mouse models, B7-H3 (Cd276) knockout (KO) in tumor cells, but not in stromal cells, suppressed tumor progression, with a reduced number of M2 macrophages and an increased number of IFNγ+CD8+ T cells. CCL2 expression was downregulated in the B7-H3 KO tumor cell lines. Inhibition of the CCL2-CCR2 axis partly negated the effects of B7-H3 suppression on M2 macrophage migration and differentiation, and tumor progression. In patients with HGSOC, B7-H3 expression positively correlated with CCL2 expression and M2 macrophage abundance, and patients with B7-H3-high tumors had fewer tumoral IFNγ+CD8+ T cells and poorer prognosis than patients with B7-H3-low tumors. Thus, B7-H3 expression in tumor cells contributes to CCL2-CCR2-M2 macrophage axis-mediated immunosuppression and tumor progression. These findings provide new insights into the immunologic TME and could aid the development of new therapeutic approaches against the unfavorable HGSOC phenotype.
Collapse
Affiliation(s)
- Taito Miyamoto
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Gynecology, Shiga General Hospital, Moriyama, Japan.,Corresponding Author: Ryusuke Murakami, Department of Gynecology, Shiga General Hospital, 5-4-30, Moriyama, Moriyama City, Shiga 524-8524, Japan. Phone: 817-7582-5031; Fax: 817-7582-5931; E-mail:
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Yuko Hosoe
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nathan Mise
- Department of Environmental and Preventive Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Shiro Takamatsu
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuka Mise
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masayo Ukita
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mana Taki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoki Horikawa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Obstetrics and Gynecology, Shizuoka General Hospital, Shizuoka, Japan
| | - Kaoru Abiko
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University School of Medicine, Higashiosaka, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
78
|
Hourani T, Holden JA, Li W, Lenzo JC, Hadjigol S, O’Brien-Simpson NM. Tumor Associated Macrophages: Origin, Recruitment, Phenotypic Diversity, and Targeting. Front Oncol 2021; 11:788365. [PMID: 34988021 PMCID: PMC8722774 DOI: 10.3389/fonc.2021.788365] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/30/2021] [Indexed: 12/20/2022] Open
Abstract
The tumor microenvironment (TME) is known to have a strong influence on tumorigenesis, with various components being involved in tumor suppression and tumor growth. A protumorigenic TME is characterized by an increased infiltration of tumor associated macrophages (TAMs), where their presence is strongly associated with tumor progression, therapy resistance, and poor survival rates. This association between the increased TAMs and poor therapeutic outcomes are stemming an increasing interest in investigating TAMs as a potential therapeutic target in cancer treatment. Prominent mechanisms in targeting TAMs include: blocking recruitment, stimulating repolarization, and depletion methods. For enhancing targeting specificity multiple nanomaterials are currently being explored for the precise delivery of chemotherapeutic cargo, including the conjugation with TAM-targeting peptides. In this paper, we provide a focused literature review of macrophage biology in relation to their role in tumorigenesis. First, we discuss the origin, recruitment mechanisms, and phenotypic diversity of TAMs based on recent investigations in the literature. Then the paper provides a detailed review on the current methods of targeting TAMs, including the use of nanomaterials as novel cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Neil M. O’Brien-Simpson
- Antimicrobial, Cancer Therapeutics and Vaccines (ACTV) Research Group, Melbourne Dental School, Centre for Oral Health Research, Royal Dental Hospital, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
79
|
Begum Y, Pandit A, Swarnakar S. Insights Into the Regulation of Gynecological Inflammation-Mediated Malignancy by Metalloproteinases. Front Cell Dev Biol 2021; 9:780510. [PMID: 34912809 PMCID: PMC8667270 DOI: 10.3389/fcell.2021.780510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/12/2021] [Indexed: 12/09/2022] Open
Abstract
Gynecological illness accounts for around 4.5% of the global disease burden, which is higher than other key global health concerns such as malaria (1.04%), TB (1.9%), ischemic heart disease (2.2%), and maternal disorders (3.5%). Gynecological conditions in women of reproductive age are linked to both in terms of diagnosis and treatment, especially in low-income economies, which poses a serious social problem. A greater understanding of health promotion and illness management can help to prevent diseases in gynecology. Due to the lack of established biomarkers, the identification of gynecological diseases, including malignancies, has proven to be challenging in most situations, and histological exams remain the gold standard. Metalloproteinases (MMPs, ADAMs, ADAMTSs) and their endogenous inhibitors (TIMPs) modulate the protease-dependent bioavailability of local niche components (e.g., growth factors), matrix turnover, and cellular interactions to govern specific physical and biochemical characteristics of the environment. Matrix metalloproteinases (MMPs), A Disintegrin and Metalloproteinase (ADAM), and A Disintegrin and Metalloproteinase with Thrombospondin Motif (ADAMTS) are zinc-dependent endopeptidases that contribute significantly to the disintegration of extracellular matrix proteins and shedding of membrane-bound receptor molecules in several diseases, including arthritis. MMPs are noteworthy genes associated with cancer development, functional angiogenesis, invasion, metastasis, and immune surveillance evasion. These genes are often elevated in cancer and multiple benign gynecological disorders like endometriosis, according to research. Migration through the extracellular matrix, which involves proteolytic activity, is an essential step in tumor cell extravasation and metastasis. However, none of the MMPs’ expression patterns, as well as their diagnostic and prognostic potential, have been studied in a pan-cancer context. The latter plays a very important role in cell signaling and might be used as a cancer treatment target. ADAMs are implicated in tumor cell proliferation, angiogenesis, and metastasis. This review will focus on the contribution of the aforementioned metalloproteinases in regulating gynecological disorders and their subsequent manipulation for therapeutic intervention.
Collapse
Affiliation(s)
- Yasmin Begum
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Anuradha Pandit
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Snehasikta Swarnakar
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
80
|
Aghlara-Fotovat S, Nash A, Kim B, Krencik R, Veiseh O. Targeting the extracellular matrix for immunomodulation: applications in drug delivery and cell therapies. Drug Deliv Transl Res 2021; 11:2394-2413. [PMID: 34176099 DOI: 10.1007/s13346-021-01018-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 12/12/2022]
Abstract
Host immune cells interact bi-directionally with their extracellular matrix (ECM) to receive and deposit molecular signals, which orchestrate cellular activation, proliferation, differentiation, and function to maintain healthy tissue homeostasis. In response to pathogens or damage, immune cells infiltrate diseased sites and synthesize critical ECM molecules such as glycoproteins, proteoglycans, and glycosaminoglycans to promote healing. When the immune system misidentifies pathogens or fails to survey damaged cells effectively, maladies such as chronic inflammation, autoimmune diseases, and cancer can develop. In these conditions, it is essential to restore balance to the body through modulation of the immune system and the ECM. This review details the components of dysregulated ECM implicated in pathogenic environments and therapeutic approaches to restore tissue homeostasis. We evaluate emerging strategies to overcome inflamed, immune inhibitory, and otherwise diseased microenvironments, including mechanical stimulation, targeted proteases, adoptive cell therapy, mechanomedicine, and biomaterial-based cell therapeutics. We highlight various strategies that have produced efficacious responses in both pre-clinical and human trials and identify additional opportunities to develop next-generation interventions. Significantly, we identify a need for therapies to address dense or fibrotic tissue for the treatment of organ tissue damage and various cancer subtypes. Finally, we conclude that therapeutic techniques that disrupt, evade, or specifically target the pathogenic microenvironment have a high potential for improving therapeutic outcomes and should be considered a priority for immediate exploration. A schematic showing the various methods of extracellular matrix disruption/targeting in both fibrotic and cancerous environments. a Biomaterial-based cell therapy can be used to deliver anti-inflammatory cytokines, chemotherapeutics, or other factors for localized, slow release of therapeutics. b Mechanotherapeutics can be used to inhibit the deposition of molecules such as collagen that affect stiffness. c Ablation of the ECM and target tissue can be accomplished via mechanical degradation such as focused ultrasound. d Proteases can be used to improve the distribution of therapies such as oncolytic virus. e Localization of therapeutics such as checkpoint inhibitors can be improved with the targeting of specific ECM components, reducing off-target effects and toxicity.
Collapse
Affiliation(s)
| | - Amanda Nash
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| |
Collapse
|
81
|
Yang C, Chen C, Xiao Q, Wang X, Shou Y, Tian X, Wang S, Li H, Liang Y, Shu J, Chen K, Sun M. Relationship Between PTEN and Angiogenesis of Esophageal Squamous Cell Carcinoma and the Underlying Mechanism. Front Oncol 2021; 11:739297. [PMID: 34796109 PMCID: PMC8593196 DOI: 10.3389/fonc.2021.739297] [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: 07/10/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) has high morbidity and mortality rates owing to its ability to infiltrate and metastasize. Microvessels formed in early-stage ESCC promote metastasis. Phosphatase and tensin homolog (PTEN) mediates macrophage polarization, but its effect and mechanism on early ESCC angiogenesis are unclear. To explore the molecular mechanism underlying early ESCC metastasis through blood vessels, we investigated the relationship between PTEN/phosphoinositide 3-kinase (PI3K)/p-AKT protein levels, number of infiltrated macrophages, and angiogenesis in ESCC and ESCC-adjacent normal esophageal mucosa tissues from 49 patients. Additionally, PTEN was overexpressed or silenced in the esophageal cancer cell line EC9706, and its supernatant served as conditioning medium for M1 tumor-associated macrophages (TAMs). The culture medium of macrophages served as conditioning medium for esophageal tumor-associated vascular endothelial cells (TECs) to study the biological behavior of PTEN-plasmid, PTEN-siRNA, and control TECs. We found that M1 TAM infiltration in ESCC tissues was low, whereas M2 TAM infiltration was high. Microvessel density was large, PTEN was down-regulated, and the PI3K/AKT pathway was activated in ESCC specimens. These parameters significantly related to the depth of tumor invasion, lymph node metastasis, and pathological staging of ESCC. Silencing of PTEN in EC9706 cells significantly activated the PI3K/AKT signaling pathway in macrophages, promoting M1-to-M2 TAM polarization and enhancing TECs’ ability to proliferate, migrate, invade, form tubes, and secrete vascular endothelial growth factor. We believe that PTEN silencing in esophageal cancer cells activates the PI3K/AKT signaling pathway in macrophages via the tumor microenvironment, induces M2 TAM polarization, and enhances the malignant behavior of TECs, thereby promoting ESCC angiogenesis. Our findings lay an empirical foundation for the development of novel diagnostic and therapeutic strategies for ESCC.
Collapse
Affiliation(s)
- Chenbo Yang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Chao Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Qiankun Xiao
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Xiaoqian Wang
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Yuwei Shou
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Xiangyu Tian
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Shuaiyuan Wang
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Hui Li
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Yinghao Liang
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Jiao Shu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Miaomiao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
82
|
Fucikova J, Coosemans A, Orsulic S, Cibula D, Vergote I, Galluzzi L, Spisek R. Immunological configuration of ovarian carcinoma: features and impact on disease outcome. J Immunother Cancer 2021; 9:jitc-2021-002873. [PMID: 34645669 PMCID: PMC8515436 DOI: 10.1136/jitc-2021-002873] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
Epithelial ovarian carcinoma (EOC) is a relatively rare malignancy but is the fifth-leading cause of cancer-related death in women, largely reflecting early, prediagnosis dissemination of malignant disease to the peritoneum. At odds with other neoplasms, EOC is virtually insensitive to immune checkpoint inhibitors, correlating with a tumor microenvironment that exhibits poor infiltration by immune cells and active immunosuppression. Here, we comparatively summarize the humoral and cellular features of primary and metastatic EOC, comparatively analyze their impact on disease outcome, and propose measures to alter them in support of treatment sensitivity and superior patient survival.
Collapse
Affiliation(s)
- Jitka Fucikova
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sandra Orsulic
- UCLA David Geffen School of Medicine and Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| | - David Cibula
- Gynecologic Oncology Center, Department of Obstetrics and Gynecology, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ignace Vergote
- Division of Gynecological Oncology, Department of Obstetrics and Gynecology, University Hospital Leuven, Leuven, Belgium
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
83
|
Pisano S, Lenna S, Healey GD, Izardi F, Meeks L, Jimenez YS, Velazquez OS, Gonzalez D, Conlan RS, Corradetti B. Assessment of the immune landscapes of advanced ovarian cancer in an optimized in vivo model. Clin Transl Med 2021; 11:e551. [PMID: 34709744 PMCID: PMC8506632 DOI: 10.1002/ctm2.551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Ovarian cancer (OC) is typically diagnosed late, associated with high rates of metastasis and the onset of ascites during late stage disease. Understanding the tumor microenvironment and how it impacts the efficacy of current treatments, including immunotherapies, needs effective in vivo models that are fully characterized. In particular, understanding the role of immune cells within the tumor and ascitic fluid could provide important insights into why OC fails to respond to immunotherapies. In this work, we comprehensively described the immune cell infiltrates in tumor nodules and the ascitic fluid within an optimized preclinical model of advanced ovarian cancer. METHODS Green Fluorescent Protein (GFP)-ID8 OC cells were injected intraperitoneally into C57BL/6 mice and the development of advanced stage OC monitored. Nine weeks after tumor injection, mice were sacrificed and tumor nodules analyzed to identify specific immune infiltrates by immunohistochemistry. Ascites, developed in tumor bearing mice over a 10-week period, was characterized by mass cytometry (CyTOF) to qualitatively and quantitatively assess the distribution of the immune cell subsets, and their relationship to ascites from ovarian cancer patients. RESULTS Tumor nodules in the peritoneal cavity proved to be enriched in T cells, antigen presenting cells and macrophages, demonstrating an active immune environment and cell-mediated immunity. Assessment of the immune landscape in the ascites showed the predominance of CD8+ , CD4+ , B- , and memory T cells, among others, and the coexistance of different immune cell types within the same tumor microenvironment. CONCLUSIONS We performed, for the first time, a multiparametric analysis of the ascitic fluid and specifically identify immune cell populations in the peritoneal cavity of mice with advanced OC. Data obtained highlights the impact of CytOF as a diagnostic tool for this malignancy, with the opportunity to concomitantly identify novel targets, and define personalized therapeutic options.
Collapse
Affiliation(s)
- Simone Pisano
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
- Center for NanoHealthSwansea University Medical SchoolSwanseaUK
| | - Stefania Lenna
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
| | | | | | - Lucille Meeks
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
| | - Yajaira S. Jimenez
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
- Texas A&M Health Science CenterCollege of MedicineBryanTexas
| | - Oscar S Velazquez
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
| | | | - Robert Steven Conlan
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
- Center for NanoHealthSwansea University Medical SchoolSwanseaUK
| | - Bruna Corradetti
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
- Center for NanoHealthSwansea University Medical SchoolSwanseaUK
- Texas A&M Health Science CenterCollege of MedicineBryanTexas
| |
Collapse
|
84
|
Wei Z, Yang M, Feng M, Wu Z, Rosin-Arbesfeld R, Dong J, Zhu D. Inhibition of BCL9 Modulates the Cellular Landscape of Tumor-Associated Macrophages in the Tumor Immune Microenvironment of Colorectal Cancer. Front Pharmacol 2021; 12:713331. [PMID: 34566638 PMCID: PMC8461101 DOI: 10.3389/fphar.2021.713331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/20/2021] [Indexed: 01/01/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are an indispensable part of the tumor microenvironment (TME), and they likely play a negative rather than positive role in cancer treatment. However, the cellular landscape and transcriptional profile regulation of TAMs in the case of tumor gene inactivation or chemical interference remains unclear. The B-cell lymphoma 9/B-cell lymphoma 9-like (BCL9/BCL9L) is a critical transcription co-factor of β-catenin. Suppression of Bcl9 inhibits tumor growth in mouse models of colorectal cancer (CRC). Here, we studied the TAMs of CRC by single-cell sequencing. Bcl9 depletion caused macrophage polarization inhibition from M0 to M2 and changed the CRC TME, which further interferes with the inflammation of M0 and M1. The transcription factor regulating these processes may be related to the Wnt signaling pathway from multiple levels. Furthermore, we also found that the cells delineated from monocyte to NK-like non-functioning cells were significantly different in the BCL9-deprived population. Combining these data, we proposed a TAM-to-NK score to evaluate the dynamic balance in TME of monocyte/TAM cells and NK-like non-functioning cells in The Cancer Genome Atlas (TCGA) clinical samples to verify the clinical significance. We demonstrated that the cell type balance and transcription differences of TAMs regulated by BCL9-driven Wnt signaling affected immune surveillance and inflammation of cancer, ultimately affecting patients' prognosis. We thereby highlighted the potential of targeting Wnt signaling pathway through cancer immunotherapy.
Collapse
Affiliation(s)
- Zhuang Wei
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Key Laboratory of Systems Biology, Innovation Center for Cell Signaling Network, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mengxuan Yang
- Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mei Feng
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Zhongen Wu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Rina Rosin-Arbesfeld
- Department of Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jibin Dong
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Di Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Key Laboratory of Smart Drug Delivery, State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Ministry of Education, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of ImmunoTherapeutics, Fudan University, Shanghai, China
| |
Collapse
|
85
|
Zheng M, Hu Y, Gou R, Li S, Nie X, Li X, Lin B. Development of a seven-gene tumor immune microenvironment prognostic signature for high-risk grade III endometrial cancer. MOLECULAR THERAPY-ONCOLYTICS 2021; 22:294-306. [PMID: 34553020 PMCID: PMC8426172 DOI: 10.1016/j.omto.2021.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/02/2021] [Indexed: 02/06/2023]
Abstract
Uterine corpus endometrial carcinoma locally infiltrates numerous immune cells and other tumor immune microenvironment components. These cells are involved in malignant tumor growth and proliferation and the process of resistance toward immunotherapies. Here, we aimed to develop a tumor immune microenvironment-related prognostic signature for high-risk grade III endometrial carcinoma based on The Cancer Genome Atlas. The signature was systematically correlated with immune infiltration characteristics of the tumor microenvironment. The seven-gene Riskscore signature was robust and performed well in training, testing, and Gene Expression Omnibus-independent cohorts. A nomogram comprising the gene signature accurately predicted patient prognosis, with our model performing better than other endometrial cancer-related signatures. Analysis of the IMvigor210 immunotherapy cohort revealed that subgroups with a low Riskscore had a better prognosis than subgroups with a high Riskscore. Subgroups with a low Riskscore exhibited immune cell infiltration and inflammatory profiles, whereas subgroups with a high Riskscore experienced progressive disease. The receiver operating characteristic curve indicated that risk score, neoantigen, and tumor mutation burden models together accurately predicted treatment response. Taken together, we developed a tumor microenvironment-based seven-gene prognostic stratification system to predict the prognosis of patients with high-risk endometrial cancer and guide more effective immunotherapy strategies.
Collapse
Affiliation(s)
- Mingjun Zheng
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China.,Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany
| | - Yuexin Hu
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| | - Rui Gou
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| | - Siting Li
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| | - Xin Nie
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| | - Xiao Li
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| | - Bei Lin
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| |
Collapse
|
86
|
Reis-Sobreiro M, Teixeira da Mota A, Jardim C, Serre K. Bringing Macrophages to the Frontline against Cancer: Current Immunotherapies Targeting Macrophages. Cells 2021; 10:2364. [PMID: 34572013 PMCID: PMC8464913 DOI: 10.3390/cells10092364] [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: 06/30/2021] [Revised: 08/07/2021] [Accepted: 08/29/2021] [Indexed: 12/21/2022] Open
Abstract
Macrophages are found in all tissues and display outstanding functional diversity. From embryo to birth and throughout adult life, they play critical roles in development, homeostasis, tissue repair, immunity, and, importantly, in the control of cancer growth. In this review, we will briefly detail the multi-functional, protumoral, and antitumoral roles of macrophages in the tumor microenvironment. Our objective is to focus on the ever-growing therapeutic opportunities, with promising preclinical and clinical results developed in recent years, to modulate the contribution of macrophages in oncologic diseases. While the majority of cancer immunotherapies target T cells, we believe that macrophages have a promising therapeutic potential as tumoricidal effectors and in mobilizing their surroundings towards antitumor immunity to efficiently limit cancer progression.
Collapse
Affiliation(s)
| | | | | | - Karine Serre
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal; (M.R.-S.); (A.T.d.M.); (C.J.)
| |
Collapse
|
87
|
Kishore A, Petrek M. Roles of Macrophage Polarization and Macrophage-Derived miRNAs in Pulmonary Fibrosis. Front Immunol 2021; 12:678457. [PMID: 34489932 PMCID: PMC8417529 DOI: 10.3389/fimmu.2021.678457] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/29/2021] [Indexed: 12/24/2022] Open
Abstract
This mini-review summarizes the current evidence for the role of macrophage activation and polarization in inflammation and immune response pertinent to interstitial lung disease, specifically pulmonary fibrosis. In the fibrosing lung, the production and function of inflammatory and fibrogenic mediators involved in the disease development have been reported to be regulated by the effects of polarized M1/M2 macrophage populations. The M1 and M2 macrophage phenotypes were suggested to correspond with the pro-inflammatory and pro-fibrogenic signatures, respectively. These responses towards tissue injury followed by the development and progression of lung fibrosis are further regulated by macrophage-derived microRNAs (miRNAs). Besides cellular miRNAs, extracellular exosomal-miRNAs derived from M2 macrophages have also been proposed to promote the progression of pulmonary fibrosis. In a future perspective, harnessing the noncoding miRNAs with a key role in the macrophage polarization is, therefore, suggested as a promising therapeutic strategy for this debilitating disease.
Collapse
Affiliation(s)
- Amit Kishore
- Department of Pathological Physiology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia.,Accuscript Consultancy, Ludhiana, India
| | - Martin Petrek
- Department of Pathological Physiology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia.,Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia.,Departments of Experimental Medicine, and Immunology, University Hospital Olomouc, Olomouc, Czechia
| |
Collapse
|
88
|
Chamseddine AN, Assi T, Mir O, Chouaib S. Modulating tumor-associated macrophages to enhance the efficacy of immune checkpoint inhibitors: A TAM-pting approach. Pharmacol Ther 2021; 231:107986. [PMID: 34481812 DOI: 10.1016/j.pharmthera.2021.107986] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAM) plasticity and diversity are both essential hallmarks of the monocyte-macrophage lineage and the tumor-derived inflammation. TAM exemplify the perfect adaptable cell with dynamic phenotypic modifications that reflect changes in their functional polarization status. Under several tumor microenvironment (TME)-related cues, TAM shift their polarization, hence promoting or halting cancer progression. Immune checkpoint inhibitors (ICI) displayed unprecedented clinical responses in various refractory cancers; but only approximately a third of patients experienced durable responses. It is, therefore, crucial to enhance the response rate of immunotherapy. Several mechanisms of resistance to ICI have been elucidated including TAM role with its essential immunosuppressive functions that reduce both anti-tumor immunity and the subsequent ICI efficacy. In the past few years, thorough research has led to a better understanding of TAM biology and innovative approaches can now be adapted through targeting macrophages' recruitment axis as well as TAM activation and polarization status within the TME. Some of these therapeutic strategies are currently being evaluated in several clinical trials in association with ICI agents. This combination between TAM modulation and ICI allows targeting TAM intrinsic immunosuppressive functions and tumor-promoting factors as well as overcoming ICI resistance. Hence, such strategies, with a better understanding of the mechanisms driving TAM modulation, may have the potential to optimize ICI efficacy.
Collapse
Affiliation(s)
- Ali N Chamseddine
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Biostatistics and Epidemiology, CESP INSERM U1018, OncoStat, Gustave Roussy, F-94805, Villejuif, France.
| | - Tarek Assi
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France
| | - Olivier Mir
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Pharmacology, Gustave Roussy, F-94805, Villejuif, France; Department of Ambulatory Care, Gustave Roussy, F-94805, Villejuif, France
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, F-94805, Villejuif, France
| |
Collapse
|
89
|
Monocyte Infiltration and Differentiation in 3D Multicellular Spheroid Cancer Models. Pathogens 2021; 10:pathogens10080969. [PMID: 34451433 PMCID: PMC8399809 DOI: 10.3390/pathogens10080969] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/01/2023] Open
Abstract
Tumor-associated macrophages often correlate with tumor progression, and therapies targeting immune cells in tumors have emerged as promising treatments. To select effective therapies, we established an in vitro 3D multicellular spheroid model including cancer cells, fibroblasts, and monocytes. We analyzed monocyte infiltration and differentiation in spheroids generated from fibroblasts and either of the cancer cell lines MCF-7, HT-29, PANC-1, or MIA PaCa-2. Monocytes rapidly infiltrated spheroids and differentiated into mature macrophages with diverse phenotypes in a cancer cell line-dependent manner. MIA PaCa-2 spheroids polarized infiltrating monocytes to M2-like macrophages with high CD206 and CD14 expression, whereas monocytes polarized by MCF-7 spheroids displayed an M1-like phenotype. Monocytes in HT-29 and PANC-1 primarily obtained an M2-like phenotype but also showed upregulation of M1 markers. Analysis of the secretion of 43 soluble factors demonstrated that the cytokine profile between spheroid cultures differed considerably depending on the cancer cell line. Secretion of most of the cytokines increased upon the addition of monocytes resulting in a more inflammatory and pro-tumorigenic environment. These multicellular spheroids can be used to recapitulate the tumor microenvironment and the phenotype of tumor-associated macrophages in vitro and provide more realistic 3D cancer models allowing the in vitro screening of immunotherapeutic compounds.
Collapse
|
90
|
Senent Y, Ajona D, González-Martín A, Pio R, Tavira B. The Complement System in Ovarian Cancer: An Underexplored Old Path. Cancers (Basel) 2021; 13:3806. [PMID: 34359708 PMCID: PMC8345190 DOI: 10.3390/cancers13153806] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer is one of the most lethal gynecological cancers. Current therapeutic strategies allow temporary control of the disease, but most patients develop resistance to treatment. Moreover, although successful in a range of solid tumors, immunotherapy has yielded only modest results in ovarian cancer. Emerging evidence underscores the relevance of the components of innate and adaptive immunity in ovarian cancer progression and response to treatment. Particularly, over the last decade, the complement system, a pillar of innate immunity, has emerged as a major regulator of the tumor microenvironment in cancer immunity. Tumor-associated complement activation may support chronic inflammation, promote an immunosuppressive microenvironment, induce angiogenesis, and activate cancer-related signaling pathways. Recent insights suggest an important role of complement effectors, such as C1q or anaphylatoxins C3a and C5a, and their receptors C3aR and C5aR1 in ovarian cancer progression. Nevertheless, the implication of these factors in different clinical contexts is still poorly understood. Detailed knowledge of the interplay between ovarian cancer cells and complement is required to develop new immunotherapy combinations and biomarkers. In this context, we discuss the possibility of targeting complement to overcome some of the hurdles encountered in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yaiza Senent
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
| | - Daniel Ajona
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Antonio González-Martín
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Oncology, Clinica Universidad de Navarra, 28027 Madrid, Spain
| | - Ruben Pio
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Beatriz Tavira
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
91
|
Sanna E, Tanca L, Cherchi C, Gramignano G, Oppi S, Chiai MG, Macciò A, Madeddu C. Decrease in Neutrophil-to-Lymphocyte Ratio during Neoadjuvant Chemotherapy as a Predictive and Prognostic Marker in Advanced Ovarian Cancer. Diagnostics (Basel) 2021; 11:1298. [PMID: 34359381 PMCID: PMC8303238 DOI: 10.3390/diagnostics11071298] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 01/04/2023] Open
Abstract
Since chronic inflammation is associated with ovarian cancer growth and progression, some clinical studies have assessed the association between the pre-treatment neutrophil-to-lymphocyte ratio (NLR) and the prognosis of ovarian cancer. The purpose of this study was to assess the dynamic behavior of the NLR during the course of neoadjuvant chemotherapy (NACT) in patients with high grade serous (HGS) advanced epithelial ovarian cancer and assess its correlation with clinical response, progression free survival (PFS) and changes in other inflammatory indexes. We performed a prospective observational study on 161 patients who underwent NACT at the Department of Gynecologic Oncology, ARNAS G. Brotzu, Cagliari, between 2009 and 2019. NLR was evaluated before starting and after three cycles of NACT. Based on response after three cycles of NACT, patients were divided into two groups: responsive and non-responsive. The primary endpoint was to assess the predictive role of NLR by comparing the responsive and non-responsive patients at baseline and after three cycles of NACT. Secondary endpoints were (a) to correlate NLR with other inflammation markers (CRP, fibrinogen, ferritin, IL-6), albumin, and modified Glasgow Prognostic Score (mGPS) with NLR at baseline and after NACT; (b) to assess the association between NLR and PFS. We found that the NLR value at baseline was not associated with response to NACT, while a decrease in NLR after three cycles was correlated with a better response to NACT. Also, values of CRP, IL-6, ferritin, and mGPS after three cycles of NACT (but not at baseline) were significantly associated with clinical response. Moreover, we found that patients with a low NLR value after 3 cycles of NACT, but not at baseline, had a significantly higher PFS than patients with high NLR after 3 cycles of NACT. In conclusion, NLR change during treatment could serve as a predictive marker of response to NACT in patients with HGS advanced ovarian cancer. This allows for the early identification of non-responsive patients who will need treatment remodeling.
Collapse
Affiliation(s)
- Elisabetta Sanna
- Department of Gynecologic Oncology, A. Businco Hospital, ARNAS G. Brotzu, 09100 Cagliari, Italy; (E.S.); (M.G.C.)
| | - Luciana Tanca
- Department of Medical Oncology, A. Businco Hospital, ARNAS G. Brotzu, 09100 Cagliari, Italy; (L.T.); (C.C.)
| | - Cristina Cherchi
- Department of Medical Oncology, A. Businco Hospital, ARNAS G. Brotzu, 09100 Cagliari, Italy; (L.T.); (C.C.)
| | - Giulia Gramignano
- Medical Oncology Unit, “Nostra Signora di Bonaria” Hospital, 09037 San Gavino, Italy;
| | - Sara Oppi
- Hematology and Transplant Center, A. Businco Hospital, ARNAS G. Brotzu, 09100 Cagliari, Italy;
| | - Maria Gloria Chiai
- Department of Gynecologic Oncology, A. Businco Hospital, ARNAS G. Brotzu, 09100 Cagliari, Italy; (E.S.); (M.G.C.)
| | - Antonio Macciò
- Department of Gynecologic Oncology, A. Businco Hospital, ARNAS G. Brotzu, 09100 Cagliari, Italy; (E.S.); (M.G.C.)
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, 09100 Cagliari, Italy;
| |
Collapse
|
92
|
Lavy M, Gauttier V, Poirier N, Barillé-Nion S, Blanquart C. Specialized Pro-Resolving Mediators Mitigate Cancer-Related Inflammation: Role of Tumor-Associated Macrophages and Therapeutic Opportunities. Front Immunol 2021; 12:702785. [PMID: 34276698 PMCID: PMC8278519 DOI: 10.3389/fimmu.2021.702785] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammation is a fundamental physiological response orchestrated by innate immune cells to restore tissue homeostasis. Specialized pro-resolving mediators (SPMs) are involved in active resolution of inflammation but when inflammation is incomplete, chronic inflammation creates a favorable environment that fuels carcinogenesis and cancer progression. Conventional cancer therapy also strengthens cancer-related inflammation by inducing massive tumor cell death that activate surrounding immune-infiltrating cells such as tumor-associated macrophages (TAMs). Macrophages are key actors of both inflammation and its active resolution due to their plastic phenotype. In line with this high plasticity, macrophages can be hijacked by cancer cells to support tumor progression and immune escape, or therapy resistance. Impaired resolution of cancer-associated inflammation supported by TAMs may thus reinforces tumor progression. From this perspective, recent evidence suggests that stimulating macrophage's pro-resolving functions using SPMs can promote inflammation resolution in cancer and improve anticancer treatments. Thus, TAMs' re-education toward an antitumor phenotype by using SPMs opens a new line of attack in cancer treatment. Here, we review SPMs' anticancer capacities with special attention regarding their effects on TAMs. We further discuss how this new therapeutic approach could be envisioned in cancer therapy.
Collapse
|
93
|
Babazadeh S, Nassiri SM, Siavashi V, Sahlabadi M, Hajinasrollah M, Zamani-Ahmadmahmudi M. Macrophage polarization by MSC-derived CXCL12 determines tumor growth. Cell Mol Biol Lett 2021; 26:30. [PMID: 34174813 PMCID: PMC8236206 DOI: 10.1186/s11658-021-00273-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Phenotypic and functional heterogeneity of macrophages is known to be the main reason for their ability to regulate inflammation and promote tumorigenesis. Mesenchymal stem cells (MSCs) are one of the principal cells commonly found in the tumor stromal niche, with capability of macrophage phenotypic switching. The objective of this study was to evaluate the role of C-X-C motif chemokine ligand 12 (CXCL12) produced by marrow-derived MSCs in the phenotypic and functional pattern of bone marrow-derived macrophages (BMDMs). METHODS First, the CRISPR/Cas9 system was used for the CXCL12 gene knock-out in MSCs. Then, coculture systems were used to investigate the role of MSCsCXCL12-/- and MSCsCXCL12+/+ in determination of macrophage phenotype. To further analyze the role of the MSC-derived CXCL12 niche, cocultures of 4T1 mammary tumor cells and macrophages primed with MSCsCXCL12-/- or MSCsCXCL12+/+ as well as in-vivo limiting dilution assays were performed. RESULTS Our results revealed that the expression of IL-4, IL-10, TGF-β and CD206 as M2 markers was significantly increased in macrophages co-cultured with MSCsCXCL12+/+ , whereas the expression of IL-6, TNF-α and iNOS was conversely decreased. The number and size of multicellular tumor spheroids were remarkably higher when 4T1 cells were cocultured with MSCCXCL12+/+-induced M2 macrophages. We also found that the occurrence of tumors was significantly higher in coinjection of 4T1 cells with MSCCXCL12+/+-primed macrophages. Tumor initiating cells were significantly decreased after coinjection of 4T1 cells with macrophages pretreated with MSCsCXCL12-/-. CONCLUSIONS In conclusion, our findings shed new light on the role of MSC-derived CXCL12 in macrophage phenotypic switching to M2, affecting their function in tumorigenesis.
Collapse
Affiliation(s)
- Shabnam Babazadeh
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Seyed Mahdi Nassiri
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Vahid Siavashi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohadeseh Sahlabadi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mostafa Hajinasrollah
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamad Zamani-Ahmadmahmudi
- Department of Clinical Science, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
94
|
Immune System-Related Changes in Preclinical GL261 Glioblastoma under TMZ Treatment: Explaining MRSI-Based Nosological Imaging Findings with RT-PCR Analyses. Cancers (Basel) 2021; 13:cancers13112663. [PMID: 34071393 PMCID: PMC8199490 DOI: 10.3390/cancers13112663] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 01/02/2023] Open
Abstract
Glioblastomas (GB) are brain tumours with poor prognosis even after aggressive therapy. Previous work suggests that magnetic resonance spectroscopic imaging (MRSI) could act as a biomarker of efficient immune system attack onto GB, presenting oscillatory changes. Glioma-associated microglia/macrophages (GAMs) constitute the most abundant non-tumour cell type within the GB and can be polarised into anti-tumour (M1) or pro-tumour (M2) phenotypes. One of the mechanisms to mediate immunosuppression in brain tumours is the interaction between programmed cell death-1 ligand 1 (PD-L1) and programmed cell death-1 receptor (PD-1). We evaluated the subpopulations of GAMs in responding and control GB tumours to correlate PD-L1 expression to GAM polarisation in order to explain/validate MRSI-detected findings. Mice were evaluated by MRI/MRSI to assess the extent of response to treatment and with qPCR for GAMs M1 and M2 polarisation analyses. M1/M2 ratios and PD-L1 expression were higher in treated compared to control tumours. Furthermore, PD-L1 expression was positively correlated with the M1/M2 ratio. The oscillatory change in the GAMs prevailing population could be one of the key causes for the differential MRSI-detected pattern, allowing this to act as immune system activity biomarker in future work.
Collapse
|
95
|
Salo H, Qu H, Mitsiou D, Aucott H, Han J, Zhang X, Aulin C, Erlandsson Harris H. Disulfide and Fully Reduced HMGB1 Induce Different Macrophage Polarization and Migration Patterns. Biomolecules 2021; 11:800. [PMID: 34071440 PMCID: PMC8229957 DOI: 10.3390/biom11060800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022] Open
Abstract
Macrophage plasticity enables cells to obtain different functions over a broad proinflammatory and repairing spectrum. In different conditions, macrophages can be induced by high-mobility group box 1 (HMGB1), a nuclear DNA-binding protein that activates innate immunity, to polarize towards a pro- (M1) or anti-inflammatory (M2) phenotype. In this study, we investigated the phenotypes of murine bone-marrow-derived macrophages (BMDMs) induced by different HMGB1 redox isoforms in depth. Our results demonstrate that disulfide HMGB1 (dsHMGB1) induces a unique macrophage phenotype that secretes pro-inflammatory cytokines, rather than inducing metabolic changes leading to nitric oxide production. Fully reduced HMGB1 (frHMGB1) did not induce macrophage polarization. The migrating function of BMDMs was measured by scratch assay after the stimulation with dsHMGB1 and frHMGB1. Both dsHMGB1 and frHMGB1 induced cell migration. We found that dsHMGB1 mediates cytokine secretion and cellular motility, mainly through toll-like receptor 4 (TLR4). Importantly, our data shows that dsHMGB1 and frHMGB1 induce distinct BMDM polarization phenotypes, and that dsHMGB1 induces a unique phenotype differing from the classical proinflammatory macrophage phenotype.
Collapse
Affiliation(s)
- Henna Salo
- Department of Medicine, Solna, Rheumatology Unit, Centre for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden; (H.S.); (H.Q.); (D.M.); (H.A.); (C.A.)
| | - Heshuang Qu
- Department of Medicine, Solna, Rheumatology Unit, Centre for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden; (H.S.); (H.Q.); (D.M.); (H.A.); (C.A.)
| | - Dimitra Mitsiou
- Department of Medicine, Solna, Rheumatology Unit, Centre for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden; (H.S.); (H.Q.); (D.M.); (H.A.); (C.A.)
| | - Hannah Aucott
- Department of Medicine, Solna, Rheumatology Unit, Centre for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden; (H.S.); (H.Q.); (D.M.); (H.A.); (C.A.)
| | - Jinming Han
- Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden; (J.H.); (X.Z.)
| | - Xingmei Zhang
- Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden; (J.H.); (X.Z.)
| | - Cecilia Aulin
- Department of Medicine, Solna, Rheumatology Unit, Centre for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden; (H.S.); (H.Q.); (D.M.); (H.A.); (C.A.)
| | - Helena Erlandsson Harris
- Department of Medicine, Solna, Rheumatology Unit, Centre for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden; (H.S.); (H.Q.); (D.M.); (H.A.); (C.A.)
| |
Collapse
|
96
|
Raskov H, Orhan A, Gaggar S, Gögenur I. Cancer-Associated Fibroblasts and Tumor-Associated Macrophages in Cancer and Cancer Immunotherapy. Front Oncol 2021; 11:668731. [PMID: 34094963 PMCID: PMC8172975 DOI: 10.3389/fonc.2021.668731] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Our understanding of the tumor microenvironment (TME), including the interplay between tumor cells, stromal cells, immune cells, and extracellular matrix components, is mandatory for the innovation of new therapeutic approaches in cancer. The cell-cell communication within the TME plays a pivotal role in the evolution and progression of cancer. Cancer-associated fibroblasts (CAF) and tumor-associated macrophages (TAM) are major cell populations in the stroma of all solid tumors and often exert protumorigenic functions; however, the origin and precise functions of CAF and TAM are still incompletely understood. CAF and TAM hold significant potential as therapeutic targets to improve outcomes in oncology when combined with existing therapies. The regulation of CAF/TAM communication and/or their differentiation could be of high impact for improving the future targeted treatment strategies. Nevertheless, there is much scope for research and innovation in this field with regards to the development of novel drugs. In this review, we elaborate on the current knowledge on CAF and TAM in cancer and cancer immunotherapy. Additionally, by focusing on their heterogenous functions in different stages and types of cancer, we explore their role as potential therapeutic targets and highlight certain aspects of their functions that need further research.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Shruti Gaggar
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
97
|
Suárez-Arriaga MC, Méndez-Tenorio A, Pérez-Koldenkova V, Fuentes-Pananá EM. Claudin-Low Breast Cancer Inflammatory Signatures Support Polarization of M1-Like Macrophages with Protumoral Activity. Cancers (Basel) 2021; 13:2248. [PMID: 34067089 PMCID: PMC8125772 DOI: 10.3390/cancers13092248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/16/2022] Open
Abstract
We previously reported that triple-negative breast cancer (BRCA) cells overexpress the cytokines GM-CSF, G-CSF, MCP-1, and RANTES, and when monocytes were 3-D co-cultured with them, M1-like macrophages were generated with the ability to induce aggressive features in luminal BRCA cell lines. These include upregulation of mesenchymal and stemness markers and invasion. In this study, we stimulated peripheral blood monocytes with the four cytokines and confirmed their capacity to generate protumoral M1-like macrophages. Using the METABRIC BRCA database, we observed that GM-CSF, MCP-1, and RANTES are associated with triple-negative BRCA and reduced overall survival, particularly in patients under 55 years of age. We propose an extended M1-like macrophage proinflammatory signature connected with these three cytokines. We found that the extended M1-like macrophage signature coexists with monocyte/macrophage, Th1 immune response, and immunosuppressive signatures, and all are enriched in claudin-low BRCA samples, and correlate with reduced patient overall survival. Furthermore, we observed that all these signatures are also present in mesenchymal carcinomas of the colon (COAD) and bladder (BLCA). The claudin-low tumor subtype has an adverse clinical outcome and remains poorly understood. This study places M1 macrophages as potential protumoral drivers in already established cancers, and as potential contributors to claudin-low aggressiveness and poor prognosis.
Collapse
Affiliation(s)
- Mayra Cecilia Suárez-Arriaga
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
- Laboratorio de Biotecnología y Bioinformática Genómica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Alfonso Méndez-Tenorio
- Laboratorio de Biotecnología y Bioinformática Genómica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Vadim Pérez-Koldenkova
- Laboratorio Nacional de Microscopía Avanzada, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Ezequiel M. Fuentes-Pananá
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
| |
Collapse
|
98
|
Li X, Liu Y, Zheng S, Zhang T, Wu J, Sun Y, Zhang J, Liu G. Role of exosomes in the immune microenvironment of ovarian cancer. Oncol Lett 2021; 21:377. [PMID: 33777201 PMCID: PMC7988709 DOI: 10.3892/ol.2021.12638] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are excretory vesicles that can deliver a variety of bioactive cargo molecules to the extracellular environment. Accumulating evidence demonstrates exosome participation in intercellular communication, immune response, inflammatory response and they even play an essential role in affecting the tumor immune microenvironment. The role of exosomes in the immune microenvironment of ovarian cancer is mainly divided into suppression and stimulation. On one hand exosomes can stimulate the innate and adaptive immune systems by activating dendritic cells (DCs), natural killer cells and T cells, allowing these immune cells exert an antitumorigenic effect. On the other hand, ovarian cancer-derived exosomes initiate cross-talk with immunosuppressive effector cells, which subsequently cause immune evasion; one of the hallmarks of cancer. Exosomes induce the polarization of macrophages in M2 phenotype and induce apoptosis of lymphocytes and DCs. Exosomes further activate additional immunosuppressive effector cells (myeloid-derived suppressor cells and regulatory T cells) that induce fibroblasts to differentiate into cancer-associated fibroblasts. Exosomes also induce the tumorigenicity of mesenchymal stem cells to exert additional immune suppression. Furthermore, besides mediating the intercellular communication, exosomes carry microRNAs (miRNAs), proteins and lipids to the tumor microenvironment, which collectively promotes ovarian cancer cells to proliferate, invade and tumors to metastasize. Studying proteins, lipids and miRNAs carried by exosomes could potentially be used as an early diagnostic marker of ovarian cancer for designing treatment strategies.
Collapse
Affiliation(s)
- Xiao Li
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yang Liu
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shuangshuang Zheng
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Tianyu Zhang
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jing Wu
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yue Sun
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jingzi Zhang
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Guoyan Liu
- Department of Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
99
|
Kim S, Han Y, Kim SI, Lee J, Jo H, Wang W, Cho U, Park WY, Rando TA, Dhanasekaran DN, Song YS. Computational modeling of malignant ascites reveals CCL5-SDC4 interaction in the immune microenvironment of ovarian cancer. Mol Carcinog 2021; 60:297-312. [PMID: 33721368 PMCID: PMC8080545 DOI: 10.1002/mc.23289] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/23/2022]
Abstract
Fluid accumulation in the abdominal cavity is commonly found in advanced-stage ovarian cancer patients, which creates a specialized tumor microenvironment for cancer progression. Using single-cell RNA sequencing (scRNA-seq) of ascites cells from five patients with ovarian cancer, we identified seven cell types, including heterogeneous macrophages and ovarian cancer cells. We resolved a distinct polarization state of macrophages by MacSpectrum analysis and observed subtype-specific enrichment of pathways associated with their functions. The communication between immune and cancer cells was predicted through a putative ligand-receptor pair analysis using NicheNet. We found that CCL5, a chemotactic ligand, is enriched in immune cells (T cells and NK cells) and mediates ovarian cancer cell survival in the ascites, possibly through SDC4. Moreover, SDC4 expression correlated with poor overall survival in ovarian cancer patients. Our study highlights the potential role of T cells and NK cells in long-term survival patients with ovarian cancer, indicating SDC4 as a potential prognostic marker in ovarian cancer patients.
Collapse
Affiliation(s)
- Soochi Kim
- Department of Neurology and Neurological Sciences, Stanford University, School of Medicine, Stanford, CA, USA
- These authors contributed equally: Soochi Kim, Youngjin Han
| | - Youngjin Han
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
- These authors contributed equally: Soochi Kim, Youngjin Han
| | - Se Ik Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Juwon Lee
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - HyunA Jo
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Wenyu Wang
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Interdisciplinary Program in Cancer Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Untack Cho
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Interdisciplinary Program in Cancer Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
- Department of Molecular Cell Biology, Sungkyunkwan University, School of Medicine, Suwon, Korea
| | - Thomas A. Rando
- Department of Neurology and Neurological Sciences, Stanford University, School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
- Neurology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Danny N. Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yong Sang Song
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Interdisciplinary Program in Cancer Biology, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
100
|
Cotechini T, Atallah A, Grossman A. Tissue-Resident and Recruited Macrophages in Primary Tumor and Metastatic Microenvironments: Potential Targets in Cancer Therapy. Cells 2021; 10:960. [PMID: 33924237 PMCID: PMC8074766 DOI: 10.3390/cells10040960] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/24/2022] Open
Abstract
Macrophages within solid tumors and metastatic sites are heterogenous populations with different developmental origins and substantially contribute to tumor progression. A number of tumor-promoting phenotypes associated with both tumor- and metastasis-associated macrophages are similar to innate programs of embryonic-derived tissue-resident macrophages. In contrast to recruited macrophages originating from marrow precursors, tissue-resident macrophages are seeded before birth and function to coordinate tissue remodeling and maintain tissue integrity and homeostasis. Both recruited and tissue-resident macrophage populations contribute to tumor growth and metastasis and are important mediators of resistance to chemotherapy, radiation therapy, and immune checkpoint blockade. Thus, targeting various macrophage populations and their tumor-promoting phenotypes holds therapeutic promise. Here, we discuss various macrophage populations as regulators of tumor progression, immunity, and immunotherapy. We provide an overview of macrophage targeting strategies, including therapeutics designed to induce macrophage depletion, impair recruitment, and induce repolarization. We also provide a perspective on the therapeutic potential for macrophage-specific acquisition of trained immunity as an anti-cancer agent and discuss the therapeutic potential of exploiting macrophages and their traits to reduce tumor burden.
Collapse
Affiliation(s)
- Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.A.); (A.G.)
| | | | | |
Collapse
|