1
|
Yang S, Zou R, Dai Y, Hu Y, Li F, Hu H. Tumor immune microenvironment and the current immunotherapy of cholangiocarcinoma (Review). Int J Oncol 2023; 63:137. [PMID: 37888583 PMCID: PMC10631767 DOI: 10.3892/ijo.2023.5585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a highly heterogeneous malignancy originating from the epithelial system of the bile ducts, and its incidence in recent years is steadily increasing. The immune microenvironment of CCA is characterized by diversity and complexity, with a substantial presence of cancer‑associated fibroblasts and immune cell infiltration, which plays a key role in regulating the distinctive biological behavior of cholangiocarcinoma, including tumor growth, angiogenesis, lymphangiogenesis, invasion and metastasis. Despite the notable success of immunotherapy in the treatment of solid tumors in recent years, patients with CCA have responded poorly to immune checkpoint inhibitor therapy. The interaction of tumor cells with cellular components of the immune microenvironment can regulate the activity and function of immune cells and form an immunosuppressive microenvironment, which may cause ineffective immunotherapy. Therefore, the components of the tumor immune microenvironment appear to be novel targets for immune therapies. Combination therapy focusing on immune checkpoint inhibitors is a promising and valuable first‑line or translational treatment approach for intractable biliary tract malignancies. The present review discusses the compositional characteristics and regulatory factors of the CCA immune microenvironment and the possible immune escape mechanisms. In addition, a summary of the advances in immunotherapy for CCA is also provided. It is hoped that the present review may function as a valuable reference for the development of novel immunotherapeutic strategies for CCA.
Collapse
Affiliation(s)
- Siqi Yang
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ruiqi Zou
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yushi Dai
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yafei Hu
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Fuyu Li
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Haijie Hu
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
2
|
Wang Y, Xu Y, Liu C, Yuan C, Zhang Y. Identification of disulfidptosis-related subgroups and prognostic signatures in lung adenocarcinoma using machine learning and experimental validation. Front Immunol 2023; 14:1233260. [PMID: 37799714 PMCID: PMC10548142 DOI: 10.3389/fimmu.2023.1233260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/04/2023] [Indexed: 10/07/2023] Open
Abstract
Background Disulfidptosis is a newly identified variant of cell death characterized by disulfide accumulation, which is independent of ATP depletion. Accordingly, the latent influence of disulfidptosis on the prognosis of lung adenocarcinoma (LUAD) patients and the progression of tumors remains poorly understood. Methods We conducted a multifaceted analysis of the transcriptional and genetic modifications in disulfidptosis regulators (DRs) specific to LUAD, followed by an evaluation of their expression configurations to define DR clusters. Harnessing the differentially expressed genes (DEGs) identified from these clusters, we formulated an optimal predictive model by amalgamating 10 distinct machine learning algorithms across 101 unique combinations to compute the disulfidptosis score (DS). Patients were subsequently stratified into high and low DS cohorts based on median DS values. We then performed an exhaustive comparison between these cohorts, focusing on somatic mutations, clinical attributes, tumor microenvironment, and treatment responsiveness. Finally, we empirically validated the biological implications of a critical gene, KYNU, through assays in LUAD cell lines. Results We identified two DR clusters and there were great differences in overall survival (OS) and tumor microenvironment. We selected the "Least Absolute Shrinkage and Selection Operator (LASSO) + Random Survival Forest (RFS)" algorithm to develop a DS based on the average C-index across different cohorts. Our model effectively stratified LUAD patients into high- and low-DS subgroups, with this latter demonstrating superior OS, a reduced mutational landscape, enhanced immune status, and increased sensitivity to immunotherapy. Notably, the predictive accuracy of DS outperformed the published LUAD signature and clinical features. Finally, we validated the DS expression using clinical samples and found that inhibiting KYNU suppressed LUAD cells proliferation, invasiveness, and migration in vitro. Conclusions The DR-based scoring system that we developed enabled accurate prognostic stratification of LUAD patients and provides important insights into the molecular mechanisms and treatment strategies for LUAD.
Collapse
Affiliation(s)
- Yuzhi Wang
- Department of Laboratory Medicine, Deyang People’s Hospital, Deyang, Sichuan, China
| | - Yunfei Xu
- Department of Laboratory Medicine, Chengdu Women’s and Children’s Central Hospital, Chengdu, Sichuan, China
| | - Chunyang Liu
- Department of Ultrasound, The First People’s Hospital of Yibin, Yibin, Sichuan, China
| | - Chengliang Yuan
- Department of Laboratory Medicine, Deyang People’s Hospital, Deyang, Sichuan, China
| | - Yi Zhang
- Department of Blood Transfusion, Deyang People’s Hospital, Deyang, Sichuan, China
| |
Collapse
|
3
|
Chen C, Ma J, Pi C, Huang W, Zhang T, Fu C, Liu W, Yang YG. PPARδ inhibition blocks the induction and function of tumor-induced IL-10 + regulatory B cells and enhances cancer immunotherapy. Cell Discov 2023; 9:54. [PMID: 37291146 DOI: 10.1038/s41421-023-00568-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023] Open
Abstract
IL-10+ regulatory B cells (Bregs) play a significant role in cancer immunotherapy and their presence is an indicator of negative outcome. We found that PPARδ is significantly upregulated in tumor-induced IL-10+ Bregs with a phenotype of CD19+CD24hiIgDlo/-CD38lo or CD19+CD24hiIgDlo/-CD38hi in both mice and humans, and the level of PPARδ expression was correlated with their potential to produce IL-10 and to inhibit T cell activation. Genetic inactivation of PPARδ in B cells impaired the development and function of IL-10+ B cells, and treatment with PPARδ inhibitor diminished the induction of IL-10+ Bregs by tumor and CD40 engagement. Importantly, immunotherapy with anti-CD40 or anti-PD1 antibody achieved a markedly improved outcome in tumor-bearing mice with PPARδ deficiency in B cells or treated with PPARδ inhibitor. This study shows that PPARδ is required for the development and function of IL-10+ Bregs, providing a new and effective target for selectively blocking Bregs and improving antitumor immunotherapy.
Collapse
Affiliation(s)
- Chen Chen
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
- Centre of Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jianan Ma
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
| | - Chenchen Pi
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
| | - Wei Huang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
| | - Tao Zhang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
| | - Cong Fu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
| | - Wentao Liu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China.
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
4
|
Macrophage Repolarization as a Therapeutic Strategy for Osteosarcoma. Int J Mol Sci 2023; 24:ijms24032858. [PMID: 36769180 PMCID: PMC9917837 DOI: 10.3390/ijms24032858] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Macrophages are versatile immune cells and can adapt to both external stimuli and their surrounding environment. Macrophages are categorized into two major categories; M1 macrophages release pro-inflammatory cytokines and produce protective responses that lead to antimicrobial or antitumor activity. M2 or tumor-associated macrophages (TAM) release anti-inflammatory cytokines that support tumor growth, invasion capacity, and metastatic potential. Since macrophages can be re-polarized from an M2 to an M1 phenotype with a variety of strategies, this has emerged as an innovative anti-cancer approach. Osteosarcoma (OS) is a kind of bone cancer and consists of a complex niche, and immunotherapy is not very effective. Therefore, immediate attention to new strategies is required. We incorporated the recent studies that have used M2-M1 repolarization strategies in the aspect of treating OS cancer.
Collapse
|
5
|
Hattinger CM, Salaroglio IC, Fantoni L, Godel M, Casotti C, Kopecka J, Scotlandi K, Ibrahim T, Riganti C, Serra M. Strategies to Overcome Resistance to Immune-Based Therapies in Osteosarcoma. Int J Mol Sci 2023; 24:ijms24010799. [PMID: 36614241 PMCID: PMC9821333 DOI: 10.3390/ijms24010799] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/14/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Improving the prognosis and cure rate of HGOSs (high-grade osteosarcomas) is an absolute need. Immune-based treatment approaches have been increasingly taken into consideration, in particular for metastatic, relapsed and refractory HGOS patients, to ameliorate the clinical results currently achieved. This review is intended to give an overview on the immunotherapeutic treatments targeting, counteracting or exploiting the different immune cell compartments that are present in the HGOS tumor microenvironment. The principle at the basis of these strategies and the possible mechanisms that HGOS cells may use to escape these treatments are presented and discussed. Finally, a list of the currently ongoing immune-based trials in HGOS is provided, together with the results that have been obtained in recently completed clinical studies. The different strategies that are presently under investigation, which are generally aimed at abrogating the immune evasion of HGOS cells, will hopefully help to indicate new treatment protocols, leading to an improvement in the prognosis of patients with this tumor.
Collapse
Affiliation(s)
- Claudia Maria Hattinger
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | | | - Leonardo Fantoni
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| | - Martina Godel
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Chiara Casotti
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
- Correspondence: (C.R.); (M.S.)
| | - Massimo Serra
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Correspondence: (C.R.); (M.S.)
| |
Collapse
|
6
|
Dendritic Cells: The Long and Evolving Road towards Successful Targetability in Cancer. Cells 2022; 11:cells11193028. [PMID: 36230990 PMCID: PMC9563837 DOI: 10.3390/cells11193028] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Dendritic cells (DCs) are a unique myeloid cell lineage that play a central role in the priming of the adaptive immune response. As such, they are an attractive target for immune oncology based therapeutic approaches. However, targeting these cells has proven challenging with many studies proving inconclusive or of no benefit in a clinical trial setting. In this review, we highlight the known and unknown about this rare but powerful immune cell. As technologies have expanded our understanding of the complexity of DC development, subsets and response features, we are now left to apply this knowledge to the design of new therapeutic strategies in cancer. We propose that utilization of these technologies through a multiomics approach will allow for an improved directed targeting of DCs in a clinical trial setting. In addition, the DC research community should consider a consensus on subset nomenclature to distinguish new subsets from functional or phenotypic changes in response to their environment.
Collapse
|
7
|
Zhang Y, He R, Lei X, Mao L, Yin Z, Zhong X, Cao W, Zheng Q, Li D. Comprehensive Analysis of a Ferroptosis-Related lncRNA Signature for Predicting Prognosis and Immune Landscape in Osteosarcoma. Front Oncol 2022; 12:880459. [PMID: 35837104 PMCID: PMC9273977 DOI: 10.3389/fonc.2022.880459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/25/2022] [Indexed: 12/19/2022] Open
Abstract
Research on the implications of ferroptosis in tumors has increased rapidly in the last decades. There are evidences that ferroptosis is involved in several aspects of cancer biology, including tumor progression, metastasis, immunomodulation, and therapeutic response. Nonetheless, the interaction between ferroptosis-related lncRNAs (FRLs) and the osteosarcoma immune microenvironment is poorly understood. In this study, a risk model composed of FRLs was developed using univariate and LASSO Cox regression analyses. On the basis of this model, FRL scores were calculated to systematically explore the role of the model in predicting the prognosis and immune characteristics of osteosarcoma patients. Survival analysis showed that osteosarcoma samples with lower FRL-score had better overall survival. After predicting the abundance of immune cells in osteosarcoma microenvironment by single-sample gene-set enrichment analysis (ssGSEA) and ESTIMATE analysis, we found that the FRL-score could distinguish immune function, immune score, stromal score, tumor purity, and tumor infiltration of immune cells in different osteosarcoma patients. In addition, FRL-score was also associated with immune checkpoint gene expression and half-maximal inhibitory concentration of chemotherapeutic agents. Finally, we confirmed that knockdown of RPARP-AS1 suppressed the malignant activity of osteosarcoma cells in vitro experiments. In general, the FRL-based prognostic signature could promote our understanding of the immune microenvironment characteristics of osteosarcoma and guide more effective treatment regimens.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Rong He
- Cancer Institute, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Xuan Lei
- Department of Burn and Plastic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lianghao Mao
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhengyu Yin
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xinyu Zhong
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wenbing Cao
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Qiping Zheng
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
- Shenzhen Academy of Peptide Targeting Technology at Pingshan, and Shenzhen Tyercan Bio-Pharm Co., Ltd., Shenzhen, China
- *Correspondence: Dapeng Li, ; Qiping Zheng,
| | - Dapeng Li
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- *Correspondence: Dapeng Li, ; Qiping Zheng,
| |
Collapse
|
8
|
Daei Sorkhabi A, Sarkesh A, Fotouhi A, Saeedi H, Aghebati-Maleki L. Cancer combination therapies by silencing of CTLA-4, PD-L1, and TIM3 in osteosarcoma. IUBMB Life 2022; 74:908-917. [PMID: 35638098 DOI: 10.1002/iub.2655] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/16/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Osteosarcoma (OS) is the most common orthopedic neoplasm, with a high metastasis rate and a dismal prognosis despite surgery and chemotherapy. Immunotherapies have offered cancer patients a ray of optimism, but their impact on OS has been disappointing. The objective of this study is to assess the effect of mono, dual, and triple combinations of CTLA-4, PD-L1, and TIM3 blockade on OS cell viability, apoptosis, and migration. METHOD The MG-63 and U-2 OS cell lines were transfected with mono, dual, and triple combinations of siRNAs specific for CTLA-4, PD-L1, and TIM3. After evaluation for transfection efficacy by qRT-PCR, MTT assay and flow cytometry were applied to assess cell viability and apoptosis rate in siRNA-transfected cells, respectively. Ultimately, the migration of transfected cells was measured by wound healing assay. RESULTS First, the qRT-PCR analysis revealed that in siRNA-transfected OS cells, CTLA-4, PD-L1, and TIM3 were downregulated. The MTT assay and flow cytometry results confirmed that silencing of these immune checkpoints in dual or triple combinations, but not in the single-agent blockade, significantly decreases cell viability and increases apoptosis, respectively. These effects were more significant when triple silencing was performed. Finally, the wound healing assay revealed that dual and triple silencing of immune checkpoints significantly inhibit cell migration, with triple silencing exhibiting a greater effect. CONCLUSION Our findings suggest that triple blockade of CTLA-4, PD-L1, and TIM3 is an effective strategy for inhibiting tumor cell progression and migration in OS, which requires large-scale clinical investigations to be translated into broad therapeutic applicability for OS patients.
Collapse
Affiliation(s)
- Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Fotouhi
- Department of Orthopedic Surgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
Discovery of New Therapeutic Targets for Osteosarcoma Treatment Based on Immune-Related lncRNAs in the Tumor Microenvironment. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3113857. [PMID: 35528175 PMCID: PMC9073548 DOI: 10.1155/2022/3113857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/22/2022] [Indexed: 11/26/2022]
Abstract
Background Long noncoding RNAs (lncRNAs) play an important role in osteosarcoma development, but their role in the tumor microenvironment (TME) is not fully understood. This study associated lncRNAs with immune-related genes and explored the mechanism of lncRNAs in osteosarcoma progression. Methods Unsupervised consensus clustering was applied to construct immune subtypes based on immune-related lncRNAs identified by Pearson's correlation analysis. A series of functional analysis was performed to reveal the links among lncRNAs, immune subtypes, TME, and osteosarcoma prognosis. Results We identified two immune subtypes C1 and C2 showing distinct overall survival. ECM-receptor interaction pathway was more activated in C2 subtype, while immune response pathways were more enriched in C2 subtype. Differential TME and response to chemotherapeutic drugs were observed between the two subtypes. Four metagenes of costimulation, cytolytic activity (CYT), immune score, and STAT1 were differentially enriched in the two subtypes. Based on 26-paired lncRNAs, we constructed a 4-paired lncRNA prognostic signature for predicting prognosis of osteosarcoma prognosis. Conclusions This study focused on immune-related lncRNAs and TME, showing the possible role and mechanisms of lncRNAs in tumor growth and metastasis. ECM may be the new therapeutic target for treating osteosarcoma, and 26-paired lncRNAs could serve as a basis for further studying the mechanisms of CYT and STAT1 in immune response, cancer cell proliferation, and migration. The two subtypes and prognostic signature could promote the design of personalized osteosarcoma treatment.
Collapse
|
10
|
Liang T, Chen J, Xu G, Zhang Z, Xue J, Zeng H, Jiang J, Chen T, Qin Z, Li H, Ye Z, Nie Y, Liu C, Zhan X. TYROBP, TLR4 and ITGAM regulated macrophages polarization and immune checkpoints expression in osteosarcoma. Sci Rep 2021; 11:19315. [PMID: 34588497 PMCID: PMC8481262 DOI: 10.1038/s41598-021-98637-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/13/2021] [Indexed: 12/22/2022] Open
Abstract
We established a relationship among the immune-related genes, tumor-infiltrating immune cells (TIICs), and immune checkpoints in patients with osteosarcoma. The gene expression data for osteosarcoma were downloaded from UCSC Xena and GEO database. Immune-related differentially expressed genes (DEGs) were detected to calculate the risk score. “Estimate” was used for immune infiltrating estimation and “xCell” was used to obtain 64 immune cell subtypes. Furthermore, the relationship among the risk scores, immune cell subtypes, and immune checkpoints was evaluated. The three immune-related genes (TYROBP, TLR4, and ITGAM) were selected to establish a risk scoring system based on their integrated prognostic relevance. The GSEA results for the Hallmark and KEGG pathways revealed that the low-risk score group exhibited the most gene sets that were related to immune-related pathways. The risk score significantly correlated with the xCell score of macrophages, M1 macrophages, and M2 macrophages, which significantly affected the prognosis of osteosarcoma. Thus, patients with low-risk scores showed better results with the immune checkpoints inhibitor therapy. A three immune-related, gene-based risk model can regulate macrophage activation and predict the treatment outcomes the survival rate in osteosarcoma.
Collapse
Affiliation(s)
- Tuo Liang
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Jiarui Chen
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - GuoYong Xu
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Zide Zhang
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Jiang Xue
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Haopeng Zeng
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Jie Jiang
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Tianyou Chen
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Zhaojie Qin
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Hao Li
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Zhen Ye
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Yunfeng Nie
- Guangxi Medical University, No.22 Shuangyong Road, Nanning, Guangxi, People's Republic of China
| | - Chong Liu
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
| | - Xinli Zhan
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China.
| |
Collapse
|
11
|
Liu Y, Feng W, Dai Y, Bao M, Yuan Z, He M, Qin Z, Liao S, He J, Huang Q, Yu Z, Zeng Y, Guo B, Huang R, Yang R, Jiang Y, Liao J, Xiao Z, Zhan X, Lin C, Xu J, Ye Y, Ma J, Wei Q, Mo Z. Single-Cell Transcriptomics Reveals the Complexity of the Tumor Microenvironment of Treatment-Naive Osteosarcoma. Front Oncol 2021; 11:709210. [PMID: 34367994 PMCID: PMC8335545 DOI: 10.3389/fonc.2021.709210] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/02/2021] [Indexed: 12/03/2022] Open
Abstract
Osteosarcoma (OS), which occurs most commonly in adolescents, is associated with a high degree of malignancy and poor prognosis. In order to develop an accurate treatment for OS, a deeper understanding of its complex tumor microenvironment (TME) is required. In the present study, tissues were isolated from six patients with OS, and then subjected to single-cell RNA sequencing (scRNA-seq) using a 10× Genomics platform. Multiplex immunofluorescence staining was subsequently used to validate the subsets identified by scRNA-seq. ScRNA-seq of six patients with OS was performed prior to neoadjuvant chemotherapy, and data were obtained on 29,278 cells. A total of nine major cell types were identified, and the single-cell transcriptional map of OS was subsequently revealed. Identified osteoblastic OS cells were divided into five subsets, and the subsets of those osteoblastic OS cells with significant prognostic correlation were determined using a deconvolution algorithm. Thereby, different transcription patterns in the cellular subtypes of osteoblastic OS cells were reported, and key transcription factors associated with survival prognosis were identified. Furthermore, the regulation of osteolysis by osteoblastic OS cells via receptor activator of nuclear factor kappa-B ligand was revealed. Furthermore, the role of osteoblastic OS cells in regulating angiogenesis through vascular endothelial growth factor-A was revealed. C3_TXNIP+ macrophages and C5_IFIT1+ macrophages were found to regulate regulatory T cells and participate in CD8+ T cell exhaustion, illustrating the possibility of immunotherapy that could target CD8+ T cells and macrophages. Our findings here show that the role of C1_osteoblastic OS cells in OS is to promote osteolysis and angiogenesis, and this is associated with survival prognosis. In addition, T cell depletion is an important feature of OS. More importantly, the present study provided a valuable resource for the in-depth study of the heterogeneity of the OS TME.
Collapse
Affiliation(s)
- Yun Liu
- Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenyu Feng
- Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yan Dai
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Mengying Bao
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Zhenchao Yuan
- Department of Bone and Soft Tissue Surgery, The Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Mingwei He
- Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhaojie Qin
- Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shijie Liao
- Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Juliang He
- Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qian Huang
- Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhenyuan Yu
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Yanyu Zeng
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Binqian Guo
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Rong Huang
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Rirong Yang
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Yonghua Jiang
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Jinling Liao
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Zengming Xiao
- Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xinli Zhan
- Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chengsen Lin
- Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Yu Ye
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Jie Ma
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qingjun Wei
- Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
12
|
Yang F, He Z, Duan H, Zhang D, Li J, Yang H, Dorsey JF, Zou W, Nabavizadeh SA, Bagley SJ, Abdullah K, Brem S, Zhang L, Xu X, Byrne KT, Vonderheide RH, Gong Y, Fan Y. Synergistic immunotherapy of glioblastoma by dual targeting of IL-6 and CD40. Nat Commun 2021; 12:3424. [PMID: 34103524 PMCID: PMC8187342 DOI: 10.1038/s41467-021-23832-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 05/17/2021] [Indexed: 01/20/2023] Open
Abstract
Immunologically-cold tumors including glioblastoma (GBM) are refractory to checkpoint blockade therapy, largely due to extensive infiltration of immunosuppressive macrophages (Mϕs). Consistent with a pro-tumor role of IL-6 in alternative Mϕs polarization, we here show that targeting IL-6 by genetic ablation or pharmacological inhibition moderately improves T-cell infiltration into GBM and enhances mouse survival; however, IL-6 inhibition does not synergize PD-1 and CTLA-4 checkpoint blockade. Interestingly, anti-IL-6 therapy reduces CD40 expression in GBM-associated Mϕs. We identify a Stat3/HIF-1α-mediated axis, through which IL-6 executes an anti-tumor role to induce CD40 expression in Mϕs. Combination of IL-6 inhibition with CD40 stimulation reverses Mϕ-mediated tumor immunosuppression, sensitizes tumors to checkpoint blockade, and extends animal survival in two syngeneic GBM models, particularly inducing complete regression of GL261 tumors after checkpoint blockade. Thus, antibody cocktail-based immunotherapy that combines checkpoint blockade with dual-targeting of IL-6 and CD40 may offer exciting opportunities for GBM and other solid tumors.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhenqiang He
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
- State Key Laboratory of Oncology in South China, Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hao Duan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
- State Key Laboratory of Oncology in South China, Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Juehui Li
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Huijuan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jay F Dorsey
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Zou
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - S Ali Nabavizadeh
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Kalil Abdullah
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Brem
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katelyn T Byrne
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert H Vonderheide
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA.
| | - Yanqing Gong
- Division of Human Genetics and Translational Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Immunotherapy for osteosarcoma: Fundamental mechanism, rationale, and recent breakthroughs. Cancer Lett 2020; 500:1-10. [PMID: 33359211 DOI: 10.1016/j.canlet.2020.12.024] [Citation(s) in RCA: 302] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Osteosarcoma (OS) is the most common primary malignancy of the bone and has a high propensity for local invasion and metastasis. Although combining surgery with chemotherapy has immensely improved the outcomes of osteosarcoma patients, the prognosis of metastatic or recurrent osteosarcomas is still unsatisfactory. Immunotherapy has proven to be a promising therapeutic strategy against human malignancies and improved understanding of the immune response to OS, and biomarker development has increased the number of patients who benefit from immunotherapies in recent years. Here, we review recent advances in immunotherapy in osteosarcoma and discuss the mechanisms and status of immunotherapies in both preclinical and clinical trials as well as future therapies on the horizon. These advances may pave the way for novel treatments requisite for patients with osteosarcoma in need of new therapies.
Collapse
|
14
|
Wang C, Li M, Wei R, Wu J. Adoptive transfer of TILs plus anti-PD1 therapy: An alternative combination therapy for treating metastatic osteosarcoma. J Bone Oncol 2020; 25:100332. [PMID: 33145154 PMCID: PMC7591383 DOI: 10.1016/j.jbo.2020.100332] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/14/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Anti-PD1 therapy for metastatic osteosarcoma patients is limited and the identification of new strategies for these patients is urgently needed. TILs plus anti-PD1 therapy significantly increases ORR, mPFS and mOS of patients. More infusion of TIL numbers and CD8+TIL percentage and less infusion of CD8+PD1+ TIL percentage and CD4+FoxP3+ TIL percentage may be potential prognostic factors which can predict clinical response to combined TILs and anti-PD1 therapy. PD1hi in fresh TILs is another good prognostic factor that predict PFS and OS.
Aim We sought to investigate the efficacy of adoptive transfer of TILs plus anti-PD1 therapy in metastatic osteosarcoma patients. Materials and methods A total of 30 patients received anti-PD1 therapy (Group 1) while 30 patients were subjected to TILs plus anti-PD1 therapy (Group 2). Progression-free survival time (PFS) and overall survival time (OS) were analyzed using Kaplan-Meier analysis. Potential prognostic factors were analyzed using univariate and multivariate analyses. Results The ORR in Group 2 is 33.3%, which is significantly higher than Group1 (6.67%). In addition, we found significantly prolonged mPFS (5.4 months) and mOS (15.2 months) in Group 2 compared to those in Group 1, which recorded mPFS and mOS of 3.8 and 6.6 months, respectively. Univariate and multivariate analyses indicate that patients with more infusions of TIL numbers and CD8+TILs or less infusions of CD8+ PD1+TILs and CD4+FoxP3+ TILs show increased PFS and OS. Moreover, PD1hi is another good prognostic factor that predict PFS and OS. Conclusion Overall, these findings indicated that TILs plus anti-PD1 therapy has significant clinical outcomes in metastatic osteosarcoma patients. However, further studies are essential to validate and characterize the therapeutic activity of TILs plus anti-PD1.
Collapse
Affiliation(s)
- Chao Wang
- Department of Orthopedic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471009, China
| | - Ming Li
- Department of Orthopedic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471009, China
| | - Rong Wei
- Department of Orthopedic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471009, China
| | - Junlong Wu
- Department of Orthopedic Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471009, China
| |
Collapse
|
15
|
Retrospective Analysis of Adoptive TIL Therapy plus Anti-PD1 Therapy in Patients with Chemotherapy-Resistant Metastatic Osteosarcoma. J Immunol Res 2020; 2020:7890985. [PMID: 33062726 PMCID: PMC7547340 DOI: 10.1155/2020/7890985] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/26/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Background The pathological subtype of osteosarcoma is one of the most common malignant bone tumors. Notably, chemotherapy-resistant metastatic osteosarcoma has been reported to cause significant mortality and shows poor prognosis with the currently available multidisciplinary treatments. This study investigated whether combined adoptive TIL and anti-PD1 therapy improves the prognosis of patients with chemotherapy-resistant metastatic osteosarcoma. Methods A total of 60 patients with chemotherapy-resistant metastatic osteosarcoma between June 2016 and March 2018 were enrolled. The primary endpoint was to evaluate the safety and adverse effects (AEs) of infusions of TIL and anti-PD1 therapy in the patients. Besides, secondary endpoints included assessing the objective response rate (ORR), progression-free survival time (PFS), and overall survival time (OS). Results We reported that combined TIL therapy and anti-PD1 therapy is safe and all treatment-related AEs were reversible or manageable. The ORR of all the patients is 36.67%, and patients with more infusions of TIL and CD8+TIL, less infusions of CD8+PD1+TIL, and less infusion of CD4+FoxP3+TIL exhibited increased PFS and OS. Conclusion This study determined that combined TIL and anti-PD1 therapy is safe and effective in metastatic osteosarcoma patients with chemotherapy resistance.
Collapse
|
16
|
Ringelstein-Harlev S. Immune dysfunction complexity in chronic lymphocytic leukemia ‒ an issue to consider when designing novel therapeutic strategies. Leuk Lymphoma 2020; 61:2050-2058. [PMID: 32336174 DOI: 10.1080/10428194.2020.1755857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
A complex interplay between chronic lymphocytic leukemia (CLL) cells and different constituents of the immune system generally results in immune tolerance. As targeted therapies are gaining a critical role in the therapeutic landscape of this disease, their impact on the already perturbed immune milieu needs to be considered. This review addresses the issues of basic immune dysfunction in CLL which is further complicated by the effects of a number of novel targeted therapies used for this malignancy. These new approaches may simultaneously facilitate both anti- and pro-cancer activity, potentially compromising the depth of response to therapy. Current evidence suggests that exploiting combination therapy could potentially overcome at least part of these deleterious effects, thereby prolonging response to treatment and helping to restore immune activity.
Collapse
Affiliation(s)
- Shimrit Ringelstein-Harlev
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
17
|
Liao P, Wang H, Tang YL, Tang YJ, Liang XH. The Common Costimulatory and Coinhibitory Signaling Molecules in Head and Neck Squamous Cell Carcinoma. Front Immunol 2019; 10:2457. [PMID: 31708918 PMCID: PMC6819372 DOI: 10.3389/fimmu.2019.02457] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/01/2019] [Indexed: 02/05/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are closely linked with immunosuppression, accompanied by complex immune cell functional activities. The abnormal competition between costimulatory and coinhibitory signal molecules plays an important role in the malignant progression of HNSCC. This review will summarize the features of costimulatory molecules (including CD137, OX40 as well as CD40) and coinhibitory molecules (including CTLA-4, PD-1, LAG3, and TIM3), analyze the underlying mechanism behind these molecules' regulation of the progression of HNSCC, and introduce the clinic application. Vaccines, such as those targeting STING while working synergistically with monoclonal antibodies, are also discussed. A deep understanding of the tumor immune landscape will help find new and improved tumor immunotherapy for HNSCC.
Collapse
Affiliation(s)
- Peng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haofan Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|