1
|
Li Z, Han B, Qi M, Li Y, Duan Y, Yao Y. Modulating macrophage-mediated programmed cell removal: An attractive strategy for cancer therapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189172. [PMID: 39151808 DOI: 10.1016/j.bbcan.2024.189172] [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: 01/11/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Macrophage-mediated programmed cell removal (PrCR) is crucial for the identification and elimination of needless cells that maintain tissue homeostasis. The efficacy of PrCR depends on the balance between pro-phagocytic "eat me" signals and anti-phagocytic "don't eat me" signals. Recently, a growing number of studies have shown that tumourigenesis and progression are closely associated with PrCR. In the tumour microenvironment, PrCR activated by the "eat me" signal is counterbalanced by the "don't eat me" signal of CD47/SIRPα, resulting in tumour immune escape. Therefore, targeting exciting "eat me" signalling while simultaneously suppressing "don't eat me" signalling and eventually inducing macrophages to produce effective PrCR will be a very attractive antitumour strategy. Here, we comprehensively review the functions of PrCR-activating signal molecules (CRT, PS, Annexin1, SLAMF7) and PrCR-inhibiting signal molecules (CD47/SIRPα, MHC-I/LILRB1, CD24/Siglec-10, SLAMF3, SLAMF4, PD-1/PD-L1, CD31, GD2, VCAM1), the interactions between these molecules, and Warburg effect. In addition, we highlight the molecular regulatory mechanisms that affect immune system function by exciting or suppressing PrCR. Finally, we review the research advances in tumour therapy by activating PrCR and discuss the challenges and potential solutions to smooth the way for tumour treatment strategies that target PrCR.
Collapse
Affiliation(s)
- Zhenzhen Li
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Bingqian Han
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Menghui Qi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yinchao Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yongtao Duan
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China.
| | - Yongfang Yao
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
2
|
Xu H, Xu D, Zheng Y, Wang H, Li A, Zheng X. Investigation of prognostic values of immune infiltration and LGMN expression in the microenvironment of osteosarcoma. Discov Oncol 2024; 15:275. [PMID: 38980440 PMCID: PMC11233489 DOI: 10.1007/s12672-024-01123-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 06/25/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Osteosarcoma (OS), the most common primary malignant bone tumor, predominantly affects children and young adults and is characterized by high invasiveness and poor prognosis. Despite therapeutic advancements, the survival rate remains suboptimal, indicating an urgent need for novel biomarkers and therapeutic targets. This study aimed to investigate the prognostic significance of LGMN expression and immune cell infiltration in the tumor microenvironment of OS. METHODS We performed an integrative bioinformatics analysis utilizing the GEO and TARGET-OS databases to identify differentially expressed genes (DEGs) associated with LGMN in OS. We conducted Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) to explore the biological pathways and functions. Additionally, we constructed protein-protein interaction (PPI) networks, a competing endogenous RNA (ceRNA) network, and applied the CIBERSORT algorithm to quantify immune cell infiltration. The diagnostic and prognostic values of LGMN were evaluated using the area under the receiver operating characteristic (ROC) curve and Cox regression analysis. Furthermore, we employed Consensus Clustering Analysis to explore the heterogeneity within OS samples based on LGMN expression. RESULTS The analysis revealed significant upregulation of LGMN in OS tissues. DEGs were enriched in immune response and antigen processing pathways, suggesting LGMN's role in immune modulation within the TME. The PPI and ceRNA network analyses provided insights into the regulatory mechanisms involving LGMN. Immune cell infiltration analysis indicated a correlation between high LGMN expression and increased abundance of M2 macrophages, implicating an immunosuppressive role. The diagnostic AUC for LGMN was 0.799, demonstrating its potential as a diagnostic biomarker. High LGMN expression correlated with reduced overall survival (OS) and progression-free survival (PFS). Importantly, Consensus Clustering Analysis identified two distinct subtypes of OS, highlighting the heterogeneity and potential for personalized medicine approaches. CONCLUSIONS Our study underscores the prognostic value of LGMN in osteosarcoma and its potential as a therapeutic target. The identification of LGMN-associated immune cell subsets and the discovery of distinct OS subtypes through Consensus Clustering Analysis provide new avenues for understanding the immunosuppressive TME of OS and may aid in the development of personalized treatment strategies. Further validation in larger cohorts is warranted to confirm these findings.
Collapse
Affiliation(s)
- Hualiang Xu
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, No. 396, Tongfu Middle Road, Haizhu District, Guangzhou, Guangdong, People's Republic of China
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, No. 613, Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, People's Republic of China
| | - Dawei Xu
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, No. 396, Tongfu Middle Road, Haizhu District, Guangzhou, Guangdong, People's Republic of China
| | - Yinfeng Zheng
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, No. 396, Tongfu Middle Road, Haizhu District, Guangzhou, Guangdong, People's Republic of China
| | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, No. 613, Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, People's Republic of China
| | - Aiguo Li
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, No. 396, Tongfu Middle Road, Haizhu District, Guangzhou, Guangdong, People's Republic of China.
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, No. 613, Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
3
|
Xiao L, Zhang L, Guo C, Xin Q, Gu X, Jiang C, Wu J. "Find Me" and "Eat Me" signals: tools to drive phagocytic processes for modulating antitumor immunity. Cancer Commun (Lond) 2024; 44:791-832. [PMID: 38923737 PMCID: PMC11260773 DOI: 10.1002/cac2.12579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Phagocytosis, a vital defense mechanism, involves the recognition and elimination of foreign substances by cells. Phagocytes, such as neutrophils and macrophages, rapidly respond to invaders; macrophages are especially important in later stages of the immune response. They detect "find me" signals to locate apoptotic cells and migrate toward them. Apoptotic cells then send "eat me" signals that are recognized by phagocytes via specific receptors. "Find me" and "eat me" signals can be strategically harnessed to modulate antitumor immunity in support of cancer therapy. These signals, such as calreticulin and phosphatidylserine, mediate potent pro-phagocytic effects, thereby promoting the engulfment of dying cells or their remnants by macrophages, neutrophils, and dendritic cells and inducing tumor cell death. This review summarizes the phagocytic "find me" and "eat me" signals, including their concepts, signaling mechanisms, involved ligands, and functions. Furthermore, we delineate the relationships between "find me" and "eat me" signaling molecules and tumors, especially the roles of these molecules in tumor initiation, progression, diagnosis, and patient prognosis. The interplay of these signals with tumor biology is elucidated, and specific approaches to modulate "find me" and "eat me" signals and enhance antitumor immunity are explored. Additionally, novel therapeutic strategies that combine "find me" and "eat me" signals to better bridge innate and adaptive immunity in the treatment of cancer patients are discussed.
Collapse
Affiliation(s)
- Lingjun Xiao
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Louqian Zhang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Ciliang Guo
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Qilei Xin
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Xiaosong Gu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| |
Collapse
|
4
|
Cao J, Qing J, Zhu L, Chen Z. Role of TIM-1 in the development and treatment of tumours. Front Cell Dev Biol 2024; 12:1307806. [PMID: 38831760 PMCID: PMC11144867 DOI: 10.3389/fcell.2024.1307806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
T-cell immunoglobulin and mucin structural domain 1 (TIM-1, also known as hepatitis A virus cell receptor 1) is a co-stimulatory molecule that is expressed predominantly on the surface of T cells. TIM-1 promotes the activation and proliferation of T cells, cytokine secretion, and can also be overexpressed in various types of cancer. Upregulation of TIM-1 expression may be associated with the development and progression of cancer. After reviewing the literature, we propose that TIM-1 affects tumour development mainly through two pathways. In the Direct pathway: overexpression in tumours activates tumour-related signaling pathways, mediates the proliferation, apoptosis, invasion and metastasis, and directly affects tumour development directly. In the indirect pathway: In addition to changing the tumour microenvironment and influencing the growth of tumours, TIM-1 binds to ligands to encourage the activation, proliferation, and generation of cytokines by immune cells. This review examines how TIM-1 stimulates the development of tumours in direct and indirect ways, and how TIM-1 is exploited as a target for cancer therapy.
Collapse
Affiliation(s)
- Jinmeng Cao
- Joint Inspection Center of Precision Medicine, The People’s Hospital of Guangxi Zhuang Autonomous Region and Guangxi Academy of Medical Sciences, Nanning, Guangxi, China
- School of Clinical Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Jilin Qing
- Center for Reproductive Medicine and Genetics, The People’s Hospital of Guangxi Zhuang Autonomous Region and Guangxi Academy of Medical Sciences, Nanning, Guangxi, China
| | - Liya Zhu
- Graduate school, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zhizhong Chen
- Joint Inspection Center of Precision Medicine, The People’s Hospital of Guangxi Zhuang Autonomous Region and Guangxi Academy of Medical Sciences, Nanning, Guangxi, China
| |
Collapse
|
5
|
Miao YR, Rankin EB, Giaccia AJ. Therapeutic targeting of the functionally elusive TAM receptor family. Nat Rev Drug Discov 2024; 23:201-217. [PMID: 38092952 PMCID: PMC11335090 DOI: 10.1038/s41573-023-00846-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 03/07/2024]
Abstract
The TAM receptor family of TYRO3, AXL and MERTK regulates tissue and immune homeostasis. Aberrant TAM receptor signalling has been linked to a range of diseases, including cancer, fibrosis and viral infections. Specifically, the dysregulation of TAM receptors can enhance tumour growth and metastasis due to their involvement in multiple oncogenic pathways. For example, TAM receptors have been implicated in the epithelial-mesenchymal transition, maintaining the stem cell phenotype, immune modulation, proliferation, angiogenesis and resistance to conventional and targeted therapies. Therapeutically, multiple TAM receptor inhibitors are in preclinical and clinical development for cancers and other indications, with those targeting AXL being the most clinically advanced. Although there has been notable clinical advancement in recent years, challenges persist. This Review aims to provide both biological and clinical insights into the current therapeutic landscape of TAM receptor inhibitors, and evaluates their potential for the treatment of cancer and non-malignant diseases.
Collapse
Affiliation(s)
- Yu Rebecca Miao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | | |
Collapse
|
6
|
Wang W, Wu S, Cen Z, Zhang Y, Chen Y, Huang Y, Cillo AR, Prokopec JS, Quarato G, Vignali DAA, Stewart-Ornstein J, Li S, Lu B, Gong YN. Mobilizing phospholipids on tumor plasma membrane implicates phosphatidylserine externalization blockade for cancer immunotherapy. Cell Rep 2022; 41:111582. [PMID: 36323258 PMCID: PMC9671066 DOI: 10.1016/j.celrep.2022.111582] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 06/06/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
In "healthy" tumor cells, phosphatidylserine (PS) is predominately localized in the inner plasma membrane leaflet. During apoptosis, PS relocates to the outer leaflet. Herein, we established PSout tumor models with tumor cells lacking PS flippase component CDC50A, constantly exposing PS but alive. PSout tumors developed bigger than wild-type (WT) tumors, featuring M2 polarized tumor-associated macrophages (TAMs) and fewer tumor-antigen-specific T cells. The PS receptor TIM-3 is responsible for PS recognition. Employing an opposite tumor model, PSin, with tumor cells lacking the PS scramblase Xkr8 and unable to expose PS during otherwise normal apoptosis, we find that the accumulated apoptotic tumor cells produce and release cyclic GAMP (cGAMP) to immune cells to activate the STING pathway, leading to TAM M1 polarization, suppressed interleukin (IL)-10 secretion, and natural killer (NK) cell cytotoxicity. Silencing Xkr8 in vivo by either short hairpin RNA (shRNA) or small interfering RNA (siRNA) to achieve a PS externalization blockade provides robust therapeutic anti-tumor efficiency.
Collapse
Affiliation(s)
- Weihong Wang
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Shaoxian Wu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Zhanpeng Cen
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; School of Medicine, Tsinghua University, Beijing, China
| | - Yixin Zhang
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Yuang Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261, USA
| | - Yixian Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261, USA
| | - Anthony R Cillo
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Joshua S Prokopec
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Giovanni Quarato
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Dario A A Vignali
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, 5115 Center Avenue, Pittsburgh, PA 15213, USA
| | - Jacob Stewart-Ornstein
- Department of Computational and Systems Biology, Hillman Cancer Center and University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, 5115 Center Avenue, Pittsburgh, PA 15213, USA.
| | - Yi-Nan Gong
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, 5115 Center Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
7
|
C3d(g), iron nanoparticles, hemin and cytochrome c may induce oxidative cytotoxicity in tumors and reduce tumor-associated myeloid cells-mediated immunosuppression. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
8
|
Lo CF, Chiu TY, Liu YT, Pan PY, Liu KL, Hsu CY, Fang MY, Huang YC, Yeh TK, Hsu TA, Chen CT, Huang LR, Tsou LK. Targeting the Phosphatidylserine-Immune Checkpoint with a Small-Molecule Maytansinoid Conjugate. J Med Chem 2022; 65:12802-12824. [PMID: 36153998 PMCID: PMC9574934 DOI: 10.1021/acs.jmedchem.2c00631] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Ligand-targeting drug delivery systems have made significant
strides
for disease treatments with numerous clinical approvals in this era
of precision medicine. Herein, we report a class of small molecule-based
immune checkpoint-targeting maytansinoid conjugates. From the ligand
targeting ability, pharmacokinetics profiling, in vivo anti-pancreatic cancer, triple-negative breast cancer, and sorafenib-resistant
liver cancer efficacies with quantitative mRNA analysis of treated-tumor
tissues, we demonstrated that conjugate 40a not only
induced lasting regression of tumor growth, but it also rejuvenated
the once immunosuppressive tumor microenvironment to an “inflamed
hot tumor” with significant elevation of gene expressions that
were not accessible in the vehicle-treated tumor. In turn, the immune
checkpoint-targeting small molecule drug conjugate from this work
represents a new pharmacodelivery strategy that can be expanded with
combination therapy with existing immune-oncology treatment options.
Collapse
Affiliation(s)
- Chen-Fu Lo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Tai-Yu Chiu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Yu-Tzu Liu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Pei-Yun Pan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Kuan-Liang Liu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Chia-Yu Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Ming-Yu Fang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Yu-Chen Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Tsu-An Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Li-Rung Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli35053, Taiwan, ROC
| |
Collapse
|
9
|
Annexin A5 as a targeting agent for cancer treatment. Cancer Lett 2022; 547:215857. [DOI: 10.1016/j.canlet.2022.215857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 11/19/2022]
|
10
|
Kaynak A, Davis HW, Kogan AB, Lee JH, Narmoneva DA, Qi X. Phosphatidylserine: The Unique Dual-Role Biomarker for Cancer Imaging and Therapy. Cancers (Basel) 2022; 14:2536. [PMID: 35626139 PMCID: PMC9139557 DOI: 10.3390/cancers14102536] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/06/2022] [Accepted: 05/17/2022] [Indexed: 01/27/2023] Open
Abstract
Cancer is among the leading causes of death worldwide. In recent years, many cancer-associated biomarkers have been identified that are used for cancer diagnosis, prognosis, screening, and early detection, as well as for predicting and monitoring carcinogenesis and therapeutic effectiveness. Phosphatidylserine (PS) is a negatively charged phospholipid which is predominantly located in the inner leaflet of the cell membrane. In many cancer cells, PS externalizes to the outer cell membrane, a process regulated by calcium-dependent flippases and scramblases. Saposin C coupled with dioleoylphosphatidylserine (SapC-DOPS) nanovesicle (BXQ-350) and bavituximab, (Tarvacin, human-mouse chimeric monoclonal antibodies) are cell surface PS-targeting drugs being tested in clinical trial for treating a variety of cancers. Additionally, a number of other PS-selective agents have been used to trigger cytotoxicity in tumor-associated endothelial cells or cancer cells in pre-clinical studies. Recent studies have demonstrated that upregulation of surface PS exposure by chemodrugs, radiation, and external electric fields can be used as a novel approach to sensitize cancer cells to PS-targeting anticancer drugs. The objectives of this review are to provide an overview of a unique dual-role of PS as a biomarker/target for cancer imaging and therapy, and to discuss PS-based anticancer strategies that are currently under active development.
Collapse
Affiliation(s)
- Ahmet Kaynak
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
| | - Harold W. Davis
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Andrei B. Kogan
- Physics Department, University of Cincinnati, Cincinnati, OH 45221, USA;
| | - Jing-Huei Lee
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
| | - Daria A. Narmoneva
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
| | - Xiaoyang Qi
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| |
Collapse
|
11
|
Stoica C, Ferreira AK, Hannan K, Bakovic M. Bilayer Forming Phospholipids as Targets for Cancer Therapy. Int J Mol Sci 2022; 23:ijms23095266. [PMID: 35563655 PMCID: PMC9100777 DOI: 10.3390/ijms23095266] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
Phospholipids represent a crucial component for the structure of cell membranes. Phosphatidylcholine and phosphatidylethanolamine are two phospholipids that comprise the majority of cell membranes. De novo biosynthesis of phosphatidylcholine and phosphatidylethanolamine occurs via the Kennedy pathway, and perturbations in the regulation of this pathway are linked to a variety of human diseases, including cancer. Altered phosphatidylcholine and phosphatidylethanolamine membrane content, phospholipid metabolite levels, and fatty acid profiles are frequently identified as hallmarks of cancer development and progression. This review summarizes the research on how phospholipid metabolism changes over oncogenic transformation, and how phospholipid profiling can differentiate between human cancer and healthy tissues, with a focus on colorectal cancer, breast cancer, and non-small cell lung cancer. The potential for phospholipids to serve as biomarkers for diagnostics, or as anticancer therapy targets, is also discussed.
Collapse
Affiliation(s)
- Celine Stoica
- Department of Human Health and Nutritional Science, College of Biological Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; (C.S.); (K.H.)
| | - Adilson Kleber Ferreira
- Department of Immunology, Laboratory of Tumor Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil;
- Department of Oncology, Alchemypet—Veterinary Dignostic Medicine, São Paulo 05024-000, Brazil
| | - Kayleigh Hannan
- Department of Human Health and Nutritional Science, College of Biological Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; (C.S.); (K.H.)
| | - Marica Bakovic
- Department of Human Health and Nutritional Science, College of Biological Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; (C.S.); (K.H.)
- Correspondence:
| |
Collapse
|
12
|
Erol A. Importance of Efferocytosis in COVID-19 Mortality. Infect Drug Resist 2022; 15:995-1007. [PMID: 35299855 PMCID: PMC8922362 DOI: 10.2147/idr.s348639] [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: 11/09/2021] [Accepted: 03/03/2022] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is a generally benign coronavirus disease that can spread rapidly, except for those with a group of risk factors. Since the pathogenesis responsible for the severity of the disease has not been clearly revealed, effective treatment alternatives has not been developed. The hallmark of the SARS-CoV-2-infected cells is apoptosis. Apoptotic cells are cleared through a sterile process defined as efferocytosis by professional and nonprofessional phagocytic cells. The disease would be rapidly brought under control in the organism that can achieve effective efferocytosis, which is also a kind of innate immune response. In the risk group, the efferocytic process is defective. With the addition of the apoptotic cell load associated with SARS-COV-2 infection, failure to achieve efferocytosis of dying cells can initiate secondary necrosis, which is a highly destructive process. Uncontrolled inflammation and coagulation abnormalities caused by secondary necrosis reason in various organ failures, lung in particular, which are responsible for the poor prognosis. Following the short and simplified information, this opinion paper aims to present possible treatment options that can control the severity of COVID-19 by detailing the mechanisms that can cause defective efferocytosis.
Collapse
Affiliation(s)
- Adnan Erol
- Independent Researcher, Not Affiliated to Any Institution, Silivri-Istanbul, Turkey
| |
Collapse
|
13
|
Immune Regulatory Processes of the Tumor Microenvironment under Malignant Conditions. Int J Mol Sci 2021; 22:ijms222413311. [PMID: 34948104 PMCID: PMC8706102 DOI: 10.3390/ijms222413311] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment (TME) is a critical regulator of tumor growth, progression, and metastasis. Since immune cells represent a large fraction of the TME, they play a key role in mediating pro- and anti-tumor immune responses. Immune escape, which suppresses anti-tumor immunity, enables tumor cells to maintain their proliferation and growth. Numerous mechanisms, which have been intensively studied in recent years, are involved in this process and based on these findings, novel immunotherapies have been successfully developed. Here, we review the composition of the TME and the mechanisms by which immune evasive processes are regulated. In detail, we describe membrane-bound and soluble factors, their regulation, and their impact on immune cell activation in the TME. Furthermore, we give an overview of the tumor/antigen presentation and how it is influenced under malignant conditions. Finally, we summarize novel TME-targeting agents, which are already in clinical trials for different tumor entities.
Collapse
|
14
|
Zhang J, Dai Z, Yan C, Wang D, Tang D. Blocking antibody-mediated phosphatidylserine enhances cancer immunotherapy. J Cancer Res Clin Oncol 2021; 147:3639-3651. [PMID: 34499223 DOI: 10.1007/s00432-021-03792-3] [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: 04/27/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy is a major breakthrough in tumor therapy and has been used in monotherapy or combination therapy. However, it has been associated with poor immune tolerance in some patients or immune-related adverse events. Therefore, ideal and reliable tumor elimination strategies are urgently needed to overcome these shortcomings. Phosphatidylserine (PS) is a negatively charged phospholipid, usually present in the inner lobules of eukaryotic cell membranes. Under certain physiological or pathological conditions, PS may be exposed on the outer leaflets of apoptotic cells serving as recognition signals by phagocytes and modulating the immune response. On the contrary, increased exposure of PS in the tumor microenvironment can significantly antagonize the body's anti-tumor immunity, thereby promoting tumor growth and metastasis. During radiotherapy and chemotherapy, PS-mediated immunosuppression increases the PS levels in necrotic tissue in the tumor microenvironment, further suppressing tumor immunity. PS-targeted therapy is a promising strategy in cancer immunotherapy. It inhibits tumor growth and improves the anti-tumor activity of immune checkpoint inhibitors. A comprehensive understanding of the mechanism of PS-targeted therapy opens up a new perspective for future cancer immunotherapies.
Collapse
Affiliation(s)
- Jie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Zhujiang Dai
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Cheng Yan
- Dalian Medical University, Dalian, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China.
| |
Collapse
|
15
|
Regulation of Tim-3 function by binding to phosphatidylserine. Biochem J 2021; 478:3999-4004. [PMID: 34813649 DOI: 10.1042/bcj20210652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/17/2022]
Abstract
Tim-3 is a transmembrane protein that is highly expressed on subsets of chronically stimulated CD4+ helper and CD8+ cytotoxic T cells, with more transient expression during acute activation and infection. Tim-3 is also constitutively expressed by multiple types of myeloid cells. Like other TIM family members, Tim-3 can bind to phosphatidylserine displayed by apoptotic cells, and this interaction has been shown to mediate uptake of such cells by dendritic cells and cross-presentation of antigens to CD8+ T cells. In contrast, how the recognition of PS by Tim-3 might regulate the function of Tim-3+ T cells is not known. In their recent paper, Lemmon and colleagues demonstrate for the first time that recognition of PS by Tim-3 leads to enhanced T cell activation.
Collapse
|
16
|
Yishake D, He TT, Liu ZY, Chen S, Luo Y, Liu XZ, Huang RZ, Lan QY, Fang AP, Zhu HL. Dietary protein and prognosis of hepatocellular carcinoma: a prospective cohort study. Food Funct 2021; 12:11568-11576. [PMID: 34709274 DOI: 10.1039/d1fo02013g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Dietary protein has been linked with all-cause and cancer mortality. However, the relationship between dietary protein and the prognosis of hepatocellular carcinoma (HCC) is still unknown. The purpose of this study was to investigate whether dietary protein intake was related to HCC mortality using data from the Guangdong Liver Cancer Cohort (GLCC), a prospective cohort study of HCC survivors established at the Sun Yat-sen University Cancer Center. Dietary information one year before the diagnosis of HCC was obtained through a 79-item semi-quantitative food frequency questionnaire (FFQ). A total of 883 patients with newly diagnosed HCC who were recruited between September 2013 and April 2017 were included in this study. The hazard ratio (HR) and 95% confidence intervals (95% CIs) were estimated by Cox proportional hazard models. The multivariate-adjusted HRs in the highest vs. the lowest tertile of total protein intake were 0.68 (95% CI: 0.52-0.91, P-trend = 0.007) for all-cause mortality and 0.74 (95% CI: 0.55-0.99, P-trend = 0.040) for HCC-specific mortality. However, the associations of animal protein intake, plant protein intake, and animal-to-plant protein ratio with all-cause and HCC-specific mortality were not significant (all P-trend >0.05). Our research suggests that higher prediagnostic dietary intake of total protein was associated with reduced all-cause and HCC-specific mortality.
Collapse
Affiliation(s)
- Dinuerguli Yishake
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.
| | - Tong-Tong He
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.
| | - Zhao-Yan Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.
| | - Si Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.
| | - Yan Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.
| | - Xiao-Zhan Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.
| | - Rong-Zhu Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.
| | - Qiu-Ye Lan
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.
| | - Ai-Ping Fang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.
| | - Hui-Lian Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.
| |
Collapse
|
17
|
Phosphatidylserine binding directly regulates TIM-3 function. Biochem J 2021; 478:3331-3349. [PMID: 34435619 PMCID: PMC8454703 DOI: 10.1042/bcj20210425] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/22/2021] [Accepted: 08/26/2021] [Indexed: 12/26/2022]
Abstract
Co-signaling receptors for the T cell receptor (TCR) are important therapeutic targets, with blockade of co-inhibitory receptors such as PD-1 now central in immuno-oncology. Advancing additional therapeutic immune modulation approaches requires understanding ligand regulation of other co-signaling receptors. One poorly understood potential therapeutic target is TIM-3 (T cell immunoglobulin and mucin domain containing-3). Which of TIM-3's several proposed regulatory ligands is/are relevant for signaling is unclear, and different studies have reported TIM-3 as a co-inhibitory or co-stimulatory receptor in T cells. Here, we show that TIM-3 promotes NF-κB signaling and IL-2 secretion following TCR stimulation in Jurkat cells, and that this activity is regulated by binding to phosphatidylserine (PS). TIM-3 signaling is stimulated by PS exposed constitutively in cultured Jurkat cells, and can be blocked by mutating the PS-binding site or by occluding this site with an antibody. We also find that TIM-3 signaling alters CD28 phosphorylation. Our findings clarify the importance of PS as a functional TIM-3 ligand, and may inform the future exploitation of TIM-3 as a therapeutic target.
Collapse
|
18
|
Hua ZD, Liu XB, Sheng JH, Li C, Li P, Cai XQ, Han ZQ. UBE2V2 Positively Correlates With PD-L1 Expression and Confers Poor Patient Survival in Lung Adenocarcinoma. Appl Immunohistochem Mol Morphol 2021; 29:585-591. [PMID: 33734107 DOI: 10.1097/pai.0000000000000928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/08/2021] [Indexed: 01/11/2023]
Abstract
This research aims to explore the diagnostic and prognostic value of ubiquitin-conjugating enzyme E2 variant 2 (UBE2V2) in lung adenocarcinoma (LUAD). The expression of UBE2V2 in clinical specimens was evaluated by bioinformatics analyses and immunohistochemistry. Bioinformatics analyses relying on the The Cancer Genome Atlas (TCGA) database suggested the elevated UBE2V2 mRNA levels in LUAD in comparison to adjacent normal tissues. Gene set enrichment analyses and gene ontology term enrichment analyses further showed the involvement of UBE2V2 in the modulation of cell cycle and immune associated signaling. The correlation analyses in TCGA LUAD data set revealed the positive correlation between UBE2V2 and CCNE1, CCNE2, CCNA2, CCNB1, CCNB2, cyclin-dependent kinase (CDK)2, CDK4, and CDK1 at the mRNA level. Moreover, UBE2V2 mRNA levels were positively correlated with PD-L1 mRNA levels, the T classification, and poor survival of LUAD patients, and were negatively correlated with type II interferon response. Consistent with the results obtained from TCGA data mining, immunohistochemistry demonstrated that UBE2V2 protein levels were upregulated in LUAD in comparison to normal tissues and were positively associated with T classification. Intriguingly, a positive correlation between UBE2V2 protein levels and PD-L1 expression was also elucidated in clinical samples. Besides, UBE2V2 expression indicated a poor prognosis in LUAD patients. Our study found that UBE2V2 was identified as an independent prognostic indicator for LUAD and might serve as an alternative target for LUAD treatment.
Collapse
Affiliation(s)
- Zhi-Dan Hua
- Department of Respiratory and Critical Care Medicine, People's Hospital of Quzhou, Quzhou, Zhejiang, China
| | | | | | | | | | | | | |
Collapse
|
19
|
Vijay R, Guthmiller JJ, Sturtz AJ, Crooks S, Johnson JT, Li L, Lan LYL, Pope RL, Chen Y, Rogers KJ, Dutta N, Toombs JE, Wilson ME, Wilson PC, Maury W, Brekken RA, Butler NS. Hemolysis-associated phosphatidylserine exposure promotes polyclonal plasmablast differentiation. J Exp Med 2021; 218:e20202359. [PMID: 33830176 PMCID: PMC8040514 DOI: 10.1084/jem.20202359] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/10/2021] [Accepted: 03/11/2021] [Indexed: 02/02/2023] Open
Abstract
Antimalarial antibody responses are essential for mediating the clearance of Plasmodium parasite-infected RBCs from infected hosts. However, the rapid appearance of large numbers of plasmablasts in Plasmodium-infected hosts can suppress the development and function of durable humoral immunity. Here, we identify that the formation of plasmablast populations in Plasmodium-infected mice is mechanistically linked to both hemolysis-induced exposure of phosphatidylserine on damaged RBCs and inflammatory cues. We also show that virus and Trypanosoma infections known to trigger hemolytic anemia and high-grade inflammation also induce exuberant plasmablast responses. The induction of hemolysis or administration of RBC membrane ghosts increases plasmablast differentiation. The phosphatidylserine receptor Axl is critical for optimal plasmablast formation, and blocking phosphatidylserine limits plasmablast expansions and reduces Plasmodium parasite burden in vivo. Our findings support that strategies aimed at modulating polyclonal B cell activation and phosphatidylserine exposure may improve immune responses against Plasmodium parasites and potentially other infectious diseases that are associated with anemia.
Collapse
Affiliation(s)
- Rahul Vijay
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA
| | - Jenna J. Guthmiller
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL
| | - Alexandria J. Sturtz
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA
| | - Sequoia Crooks
- Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, IA
| | - Jordan T. Johnson
- Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, IA
| | - Lei Li
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL
| | | | | | - Yani Chen
- Department of Internal Medicine, The University of Iowa, Iowa City, IA
| | - Kai J. Rogers
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA
| | - Nirmal Dutta
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA
| | - Jason E. Toombs
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX
| | - Mary E. Wilson
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA
- Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, IA
- Department of Internal Medicine, The University of Iowa, Iowa City, IA
- Department of Veterans Affairs Medical Center, Iowa City, IA
| | - Patrick C. Wilson
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL
- Committee on Immunology, The University of Chicago, Chicago, IL
| | - Wendy Maury
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA
- Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, IA
| | - Rolf A. Brekken
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX
- Departments of Surgery and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Noah S. Butler
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA
- Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, IA
| |
Collapse
|
20
|
Chen LB, Zhu SP, Liu TP, Zhao H, Chen PF, Duan YJ, Hu R. Cancer Associated Fibroblasts Promote Renal Cancer Progression Through a TDO/Kyn/AhR Dependent Signaling Pathway. Front Oncol 2021; 11:628821. [PMID: 33842334 PMCID: PMC8027476 DOI: 10.3389/fonc.2021.628821] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer associated fibroblasts (CAFs) play crucial roles in cancer development, however, the specific mechanisms of CAFs associated renal cancer progression remain poorly understood. Our study observed enriched CAFs in high degree malignant tumor tissues from renal cancer patients. These CAFs isolated from tumor tissues are prone to facilitate drugs resistance and promote tumor progression in vitro and in vivo. Mechanistically, CAFs up-regulated tryptophan 2, 3-dioxygenase (TDO) expression, resulting in enhanced secretion of kynurenine (Kyn). Kyn produced from CAFs could up-regulated the expression of aromatic hydrocarbon receptor (AhR), eventually resulting in the AKT and STAT3 signaling pathways activation. Inhibition of AKT signal prevented cancer cells proliferation, while inhibition of the STAT3 signal reverted drugs resistance and cancer migration induced by kynurenine. Application of AhR inhibitor DMF could efficiently suppress distant metastasis of renal cancer cells, and improve anticancer effects of sorafenib (Sor)/sunitinib (Sun), which described a promising therapeutic strategy for clinical renal cancer.
Collapse
Affiliation(s)
- Li-Bo Chen
- Department of Urology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Shun-Ping Zhu
- Department of Respiratory, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Tian-Pei Liu
- Department of Urology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Heng Zhao
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Ping-Feng Chen
- Department of Urology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - You-Jun Duan
- Department of Urology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Rong Hu
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
21
|
Marin-Acevedo JA, Kimbrough EO, Lou Y. Next generation of immune checkpoint inhibitors and beyond. J Hematol Oncol 2021; 14:45. [PMID: 33741032 PMCID: PMC7977302 DOI: 10.1186/s13045-021-01056-8] [Citation(s) in RCA: 310] [Impact Index Per Article: 103.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/05/2021] [Indexed: 12/17/2022] Open
Abstract
The immune system is the core defense against cancer development and progression. Failure of the immune system to recognize and eliminate malignant cells plays an important role in the pathogenesis of cancer. Tumor cells evade immune recognition, in part, due to the immunosuppressive features of the tumor microenvironment. Immunotherapy augments the host immune system to generate an antitumor effect. Immune checkpoints are pathways with inhibitory or stimulatory features that maintain self-tolerance and assist with immune response. The most well-described checkpoints are inhibitory in nature and include the cytotoxic T lymphocyte-associated molecule-4 (CTLA-4), programmed cell death receptor-1 (PD-1), and programmed cell death ligand-1 (PD-L1). Molecules that block these pathways to enhance the host immunologic activity against tumors have been developed and become standard of care in the treatment of many malignancies. Only a small percentage of patients have meaningful responses to these treatments, however. New pathways and molecules are being explored in an attempt to improve responses and application of immune checkpoint inhibition therapy. In this review, we aim to elucidate these novel immune inhibitory pathways, potential therapeutic molecules that are under development, and outline particular advantages and challenges with the use of each one of them.
Collapse
Affiliation(s)
| | - ErinMarie O Kimbrough
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
22
|
Davra V, Kumar S, Geng K, Calianese D, Mehta D, Gadiyar V, Kasikara C, Lahey KC, Chang YJ, Wichroski M, Gao C, De Lorenzo MS, Kotenko SV, Bergsbaken T, Mishra PK, Gause WC, Quigley M, Spires TE, Birge RB. Axl and Mertk Receptors Cooperate to Promote Breast Cancer Progression by Combined Oncogenic Signaling and Evasion of Host Antitumor Immunity. Cancer Res 2021; 81:698-712. [PMID: 33239426 PMCID: PMC9999365 DOI: 10.1158/0008-5472.can-20-2066] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/23/2020] [Accepted: 11/19/2020] [Indexed: 11/16/2022]
Abstract
Despite the promising clinical benefit of targeted and immune checkpoint blocking therapeutics, current strategies have limited success in breast cancer, indicating that additional inhibitory pathways are required to complement existing therapeutics. TAM receptors (Tyro-3, Axl, and Mertk) are often correlated with poor prognosis because of their capacities to sustain an immunosuppressive environment. Here, we ablate Axl on tumor cells using CRISPR/Cas9 gene editing, and by targeting Mertk in the tumor microenvironment (TME), we observed distinct functions of TAM as oncogenic kinases, as well as inhibitory immune receptors. Depletion of Axl suppressed cell intrinsic oncogenic properties, decreased tumor growth, reduced the incidence of lung metastasis and increased overall survival of mice when injected into mammary fat pad of syngeneic mice, and demonstrated synergy when combined with anti-PD-1 therapy. Blockade of Mertk function on macrophages decreased efferocytosis, altered the cytokine milieu, and resulted in suppressed macrophage gene expression patterns. Mertk-knockout mice or treatment with anti-Mertk-neutralizing mAb also altered the cellular immune profile, resulting in a more inflamed tumor environment with enhanced T-cell infiltration into tumors and T-cell-mediated cytotoxicity. The antitumor activity from Mertk inhibition was abrogated by depletion of cytotoxic CD8α T cells by using anti-CD8α mAb or by transplantation of tumor cells into B6.CB17-Prkdc SCID mice. Our data indicate that targeting Axl expressed on tumor cells and Mertk in the TME is predicted to have a combinatorial benefit to enhance current immunotherapies and that Axl and Mertk have distinct functional activities that impair host antitumor response. SIGNIFICANCE: This study demonstrates how TAM receptors act both as oncogenic tyrosine kinases and as receptors that mediate immune evasion in cancer progression.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cells, Cultured
- Female
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immune Evasion/genetics
- Immune Evasion/immunology
- Immunotherapy/methods
- Kaplan-Meier Estimate
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/immunology
- Proto-Oncogene Proteins/metabolism
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/immunology
- Receptor Protein-Tyrosine Kinases/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- c-Mer Tyrosine Kinase/genetics
- c-Mer Tyrosine Kinase/immunology
- c-Mer Tyrosine Kinase/metabolism
- Axl Receptor Tyrosine Kinase
- Mice
Collapse
Affiliation(s)
- Viralkumar Davra
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Sushil Kumar
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Ke Geng
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - David Calianese
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Dhriti Mehta
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Varsha Gadiyar
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Canan Kasikara
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Kevin C Lahey
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Yun-Juan Chang
- Office of Advanced Research Computing, Rutgers- New Jersey Medical School, Newark, New Jersey
| | | | - Chan Gao
- Bristol Myers Squibb, Lawrenceville, New Jersey
| | | | - Sergei V Kotenko
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Tessa Bergsbaken
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Pankaj K Mishra
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - William C Gause
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | | | | | - Raymond B Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey.
| |
Collapse
|
23
|
Gadiyar V, Lahey KC, Calianese D, Devoe C, Mehta D, Bono K, Desind S, Davra V, Birge RB. Cell Death in the Tumor Microenvironment: Implications for Cancer Immunotherapy. Cells 2020; 9:cells9102207. [PMID: 33003477 PMCID: PMC7599747 DOI: 10.3390/cells9102207] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/26/2020] [Accepted: 09/26/2020] [Indexed: 02/06/2023] Open
Abstract
The physiological fate of cells that die by apoptosis is their prompt and efficient removal by efferocytosis. During these processes, apoptotic cells release intracellular constituents that include purine nucleotides, lysophosphatidylcholine (LPC), and Sphingosine-1-phosphate (S1P) that induce migration and chemo-attraction of phagocytes as well as mitogens and extracellular membrane-bound vesicles that contribute to apoptosis-induced compensatory proliferation and alteration of the extracellular matrix and the vascular network. Additionally, during efferocytosis, phagocytic cells produce a number of anti-inflammatory and resolving factors, and, together with apoptotic cells, efferocytic events have a homeostatic function that regulates tissue repair. These homeostatic functions are dysregulated in cancers, where, aforementioned events, if not properly controlled, can lead to cancer progression and immune escape. Here, we summarize evidence that apoptosis and efferocytosis are exploited in cancer, as well as discuss current translation and clinical efforts to harness signals from dying cells into therapeutic strategies.
Collapse
|
24
|
Chang W, Fa H, Xiao D, Wang J. Targeting phosphatidylserine for Cancer therapy: prospects and challenges. Theranostics 2020; 10:9214-9229. [PMID: 32802188 PMCID: PMC7415799 DOI: 10.7150/thno.45125] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is a leading cause of mortality and morbidity worldwide. Despite major improvements in current therapeutic methods, ideal therapeutic strategies for improved tumor elimination are still lacking. Recently, immunotherapy has attracted much attention, and many immune-active agents have been approved for clinical use alone or in combination with other cancer drugs. However, some patients have a poor response to these agents. New agents and strategies are needed to overcome such deficiencies. Phosphatidylserine (PS) is an essential component of bilayer cell membranes and is normally present in the inner leaflet. In the physiological state, PS exposure on the external leaflet not only acts as an engulfment signal for phagocytosis in apoptotic cells but also participates in blood coagulation, myoblast fusion and immune regulation in nonapoptotic cells. In the tumor microenvironment, PS exposure is significantly increased on the surface of tumor cells or tumor cell-derived microvesicles, which have innate immunosuppressive properties and facilitate tumor growth and metastasis. To date, agents targeting PS have been developed, some of which are under investigation in clinical trials as combination drugs for various cancers. However, controversial results are emerging in laboratory research as well as in clinical trials, and the efficiency of PS-targeting agents remains uncertain. In this review, we summarize recent progress in our understanding of the physiological and pathological roles of PS, with a focus on immune suppressive features. In addition, we discuss current drug developments that are based on PS-targeting strategies in both experimental and clinical studies. We hope to provide a future research direction for the development of new agents for cancer therapy.
Collapse
Affiliation(s)
- Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
| | - Hongge Fa
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| | - Dandan Xiao
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| | - Jianxun Wang
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| |
Collapse
|
25
|
Calianese DC, Birge RB. Biology of phosphatidylserine (PS): basic physiology and implications in immunology, infectious disease, and cancer. Cell Commun Signal 2020; 18:41. [PMID: 32160904 PMCID: PMC7065380 DOI: 10.1186/s12964-020-00543-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Phosphatidylserine (PS) is an anionic phospholipid found on the membranes of a variety of organelles throughout the cell, most notably the plasma membrane. Under homeostatic conditions, PS is typically restricted to the inner leaflet of the plasma membrane. However, during cellular activation and/or induction of cell death, PS is externalized on the outer surface via the activation of phospholipid scramblases. Externalized PS not only changes the biochemical and biophysical properties of the plasma membrane but also initiates a series of interactions between endogenous extracellular proteins as well as receptors on neighboring cells to stimulate engulfment (efferocytosis) that influence the surrounding immune milieu. In this thematic series published in Cell Communication and Signaling, we feature review articles that highlight recent work in the field of PS biology, including the biochemistry and physiological significance of PS externalization, therapeutic applications and efforts to target PS, as well as posit open questions that remain in the field.
Collapse
Affiliation(s)
- David C Calianese
- Rutgers University, The State University of New Jersey, Newark, New Jersey, USA
| | - Raymond B Birge
- Rutgers University, The State University of New Jersey, Newark, New Jersey, USA.
| |
Collapse
|
26
|
Ghosh Roy S. TAM receptors: A phosphatidylserine receptor family and its implications in viral infections. TAM RECEPTORS IN HEALTH AND DISEASE 2020; 357:81-122. [DOI: 10.1016/bs.ircmb.2020.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|