1
|
Ricci AD, Rizzo A, Schirizzi A, D’Alessandro R, Frega G, Brandi G, Shahini E, Cozzolongo R, Lotesoriere C, Giannelli G. Tumor Immune Microenvironment in Intrahepatic Cholangiocarcinoma: Regulatory Mechanisms, Functions, and Therapeutic Implications. Cancers (Basel) 2024; 16:3542. [PMID: 39456636 PMCID: PMC11505966 DOI: 10.3390/cancers16203542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/06/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Treatment options for intrahepatic cholangiocarcinoma (iCCA), a highly malignant tumor with poor prognosis, are limited. Recent developments in immunotherapy and immune checkpoint inhibitors (ICIs) have offered new hope for treating iCCA. However, several issues remain, including the identification of reliable biomarkers of response to ICIs and immune-based combinations. Tumor immune microenvironment (TIME) of these hepatobiliary tumors has been evaluated and is under assessment in this setting in order to boost the efficacy of ICIs and to convert these immunologically "cold" tumors to "hot" tumors. Herein, the review TIME of ICCA and its critical function in immunotherapy. Moreover, this paper also discusses potential avenues for future research, including novel targets for immunotherapy and emerging treatment plans aimed to increase the effectiveness of immunotherapy and survival rates for iCCA patients.
Collapse
Affiliation(s)
- Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Rosalba D’Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Giorgio Frega
- Osteoncology, Soft Tissue and Bone Sarcomas, Innovative Therapy Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy
| | - Raffaele Cozzolongo
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| |
Collapse
|
2
|
Chen J, Duan Y, Che J, Zhu J. Dysfunction of dendritic cells in tumor microenvironment and immunotherapy. Cancer Commun (Lond) 2024; 44:1047-1070. [PMID: 39051512 PMCID: PMC11492303 DOI: 10.1002/cac2.12596] [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: 01/31/2024] [Revised: 06/10/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024] Open
Abstract
Dendritic cells (DCs) comprise diverse cell populations that play critical roles in antigen presentation and triggering immune responses in the body. However, several factors impair the immune function of DCs and may promote immune evasion in cancer. Understanding the mechanism of DC dysfunction and the diverse functions of heterogeneous DCs in the tumor microenvironment (TME) is critical for designing effective strategies for cancer immunotherapy. Clinical applications targeting DCs summarized in this report aim to improve immune infiltration and enhance the biological function of DCs to modulate the TME to prevent cancer cells from evading the immune system. Herein, factors in the TME that induce DC dysfunction, such as cytokines, hypoxic environment, tumor exosomes and metabolites, and co-inhibitory molecules, have been described. Furthermore, several key signaling pathways involved in DC dysfunction and signal-relevant drugs evaluated in clinical trials were identified. Finally, this review provides an overview of current clinical immunotherapies targeting DCs, especially therapies with proven clinical outcomes, and explores future developments in DC immunotherapies.
Collapse
Affiliation(s)
- Jie Chen
- Jecho Institute Co., LtdShanghaiP. R. China
| | - Yuhang Duan
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of EducationBeijingP. R. China
- Shanghai Jiao Tong University, School of PharmacyShanghaiP. R. China
| | - Junye Che
- Jecho Institute Co., LtdShanghaiP. R. China
| | - Jianwei Zhu
- Jecho Institute Co., LtdShanghaiP. R. China
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of EducationBeijingP. R. China
- Shanghai Jiao Tong University, School of PharmacyShanghaiP. R. China
| |
Collapse
|
3
|
Li C, Hou Y, He M, Lv L, Zhang Y, Sun S, Zhao Y, Liu X, Ma P, Wang X, Zhou Q, Zhan L. Laponite Lights Calcium Flickers by Reprogramming Lysosomes to Steer DC Migration for An Effective Antiviral CD8 + T-Cell Response. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303006. [PMID: 37638719 PMCID: PMC10602536 DOI: 10.1002/advs.202303006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/13/2023] [Indexed: 08/29/2023]
Abstract
Immunotherapy using dendritic cell (DC)-based vaccination is an established approach for treating cancer and infectious diseases; however, its efficacy is limited. Therefore, targeting the restricted migratory capacity of the DCs may enhance their therapeutic efficacy. In this study, the effect of laponite (Lap) on DCs, which can be internalized into lysosomes and induce cytoskeletal reorganization via the lysosomal reprogramming-calcium flicker axis, is evaluated, and it is found that Lap dramatically improves the in vivo homing ability of these DCs to lymphoid tissues. In addition, Lap improves antigen cross-presentation by DCs and increases DC-T-cell synapse formation, resulting in enhanced antigen-specific CD8+ T-cell activation. Furthermore, a Lap-modified cocktail (Lap@cytokine cocktail [C-C]) is constructed based on the gold standard, C-C, as an adjuvant for DC vaccines. Lap@C-C-adjuvanted DCs initiated a robust cytotoxic T-cell immune response against hepatitis B infection, resulting in > 99.6% clearance of viral DNA and successful hepatitis B surface antigen seroconversion. These findings highlight the potential value of Lap as a DC vaccine adjuvant that can regulate DC homing, and provide a basis for the development of effective DC vaccines.
Collapse
Affiliation(s)
- Chenyan Li
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
- BGI college, Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Yangyang Hou
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Minwei He
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Liping Lv
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Yulong Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Sujing Sun
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Yan Zhao
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Xingzhao Liu
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Ping Ma
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Xiaohui Wang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Qianqian Zhou
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Linsheng Zhan
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
- BGI college, Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| |
Collapse
|
4
|
Ding L, Liang M, Li Y, Zeng M, Liu M, Ma W, Chen F, Li C, Reis RL, Li F, Wang Y. Zinc-Organometallic Framework Vaccine Controlled-Release Zn 2+ Regulates Tumor Extracellular Matrix Degradation Potentiate Efficacy of Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302967. [PMID: 37439462 PMCID: PMC10520680 DOI: 10.1002/advs.202302967] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Indexed: 07/14/2023]
Abstract
Tumor extracellular matrix (ECM) not only forms a physical barrier for T cells infiltration, but also regulates multiple immunosuppressive pathways, which is an important reason for immunotherapy failure. The cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway plays a key role in activating CD8+ T cells, maintaining CD8+ T cells stemness and enhancing the antitumor effect. Herein, a zinc-organometallic framework vaccine (ZPM@OVA-CpG) prepared by self-assembly, which achieves site-directed release of Zn2+ in dendritic cell (DC) lysosomes and tumor microenvironment under acidic conditions, is reported. The vaccine actively targets DC, significantly enhances cGAS-STING signal, promotes DC maturation and antigen cross-presentation, and induces strong activation of CD8+ T cells. Meanwhile, the vaccine reaches the tumor site, releasing Zn2+ , significantly up-regulates the activity of matrix metalloproteinase-2, degrades various collagen components of tumor ECM, effectively alleviates immune suppression, and significantly enhances the tumor infiltration and killing of CD8+ T cells. ZPM@OVA-CpG vaccine not only solves the problem of low antigen delivery efficiency and weak CD8+ T cells activation ability, but also achieves the degradation of tumor ECM via the vaccine for the first time, providing a promising therapeutic platform for the development of efficient novel tumor vaccines.
Collapse
Affiliation(s)
- Lin Ding
- The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518055China
- Translational Medicine Collaborative Innovation CenterThe First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518020China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell TherapyShenzhen Key Laboratory of Stem Cell Research and Clinical TransformationShenzhen Immune Cell Therapy Public Service PlatformShenzhen518020China
| | - Minli Liang
- The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518055China
- Translational Medicine Collaborative Innovation CenterThe First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518020China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell TherapyShenzhen Key Laboratory of Stem Cell Research and Clinical TransformationShenzhen Immune Cell Therapy Public Service PlatformShenzhen518020China
| | - Yuanyuan Li
- Clinical Laboratory, Jiaozuo Women's and Child's HospitalJiaozuo454001China
| | - Mei Zeng
- School of PharmacyGuangdong Medical UniversityDongguan523109China
| | - Meiting Liu
- School of PharmacyGuangdong Medical UniversityDongguan523109China
| | - Wei Ma
- Translational Medicine Collaborative Innovation CenterThe First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518020China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell TherapyShenzhen Key Laboratory of Stem Cell Research and Clinical TransformationShenzhen Immune Cell Therapy Public Service PlatformShenzhen518020China
| | - Fuming Chen
- Translational Medicine Collaborative Innovation CenterThe First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518020China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell TherapyShenzhen Key Laboratory of Stem Cell Research and Clinical TransformationShenzhen Immune Cell Therapy Public Service PlatformShenzhen518020China
| | - Chenchen Li
- Key Laboratory of Tropical Translational Medicine of Ministry of EducationSchool of Pharmacy and The First Affiliated HospitalHainan Medical UniversityHaikou570228China
| | - Rui L. Reis
- 3B's Research GroupI3Bs‐Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoGuimarães4805–017Portugal
| | - Fu‐Rong Li
- The First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518055China
- Translational Medicine Collaborative Innovation CenterThe First Affiliated Hospital (Shenzhen People's Hospital)Southern University of Science and TechnologyShenzhen518020China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell TherapyShenzhen Key Laboratory of Stem Cell Research and Clinical TransformationShenzhen Immune Cell Therapy Public Service PlatformShenzhen518020China
| | - Yanli Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of EducationSchool of Pharmacy and The First Affiliated HospitalHainan Medical UniversityHaikou570228China
| |
Collapse
|
5
|
Khan NA, Asim M, Biswas KH, Alansari AN, Saman H, Sarwar MZ, Osmonaliev K, Uddin S. Exosome nanovesicles as potential biomarkers and immune checkpoint signaling modulators in lung cancer microenvironment: recent advances and emerging concepts. J Exp Clin Cancer Res 2023; 42:221. [PMID: 37641132 PMCID: PMC10463467 DOI: 10.1186/s13046-023-02753-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/08/2023] [Indexed: 08/31/2023] Open
Abstract
Lung cancer remains the leading cause of cancer-related deaths globally, and the survival rate remains low despite advances in diagnosis and treatment. The progression of lung cancer is a multifaceted and dynamic phenomenon that encompasses interplays among cancerous cells and their microenvironment, which incorporates immune cells. Exosomes, which are small membrane-bound vesicles, are released by numerous cell types in normal and stressful situations to allow communication between cells. Tumor-derived exosomes (TEXs) possess diverse neo-antigens and cargoes such as proteins, RNA, and DNA and have a unique molecular makeup reflecting tumor genetic complexity. TEXs contain both immunosuppressive and immunostimulatory factors and may play a role in immunomodulation by influencing innate and adaptive immune components. Moreover, they transmit signals that contribute to the progression of lung cancer by promoting metastasis, epithelial-mesenchymal transition (EMT), angiogenesis, and immunosuppression. This makes them a valuable resource for investigating the immune environment of tumors, which could pave the way for the development of non-invasive biomarkers that could aid in the prognosis, diagnosis, and immunotherapy of lung cancer. While immune checkpoint inhibitor (ICI) immunotherapy has shown promising results in treating initial-stage cancers, most patients eventually develop adaptive resistance over time. Emerging evidence demonstrates that TEXs could serve as a prognostic biomarker for immunotherapeutic response and have a significant impact on both systemic immune suppression and tumor advancement. Therefore, understanding TEXs and their role in lung cancer tumorigenesis and their response to immunotherapies is an exciting research area and needs further investigation. This review highlights the role of TEXs as key contributors to the advancement of lung cancer and their clinical significance in lung immune-oncology, including their possible use as biomarkers for monitoring disease progression and prognosis, as well as emerging shreds of evidence regarding the possibility of using exosomes as targets to improve lung cancer therapy.
Collapse
Affiliation(s)
- Naushad Ahmad Khan
- Department of Surgery, Trauma and Vascular Surgery Clinical Research, Hamad General Hospital, 3050, Doha, Qatar.
- Faculty of Medical Sciences, Ala-Too International University, Bishkek, Kyrgyzstan.
| | - Mohammad Asim
- Department of Surgery, Trauma and Vascular Surgery Clinical Research, Hamad General Hospital, 3050, Doha, Qatar
| | - Kabir H Biswas
- Division of Biological and Biomedical Sciences, College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Amani N Alansari
- Department of Surgery, Trauma and Vascular Surgery Clinical Research, Hamad General Hospital, 3050, Doha, Qatar
| | - Harman Saman
- Department of Medicine, Hazm Maubrairek Hospital, Al-Rayyan, Doha, 3050, Qatar
| | | | | | - Shahab Uddin
- Translational Research Institute & Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar.
- Department of Biosciences, Integral University, Lucknow, 226026, UP, India.
| |
Collapse
|
6
|
Huang C, Ren S, Chen Y, Liu A, Wu Q, Jiang T, Lv P, Song D, Hu F, Lan J, Sun L, Zheng X, Luo X, Chu Q, Jia K, Li Y, Wang J, Zou C, Hu J, Wang G. PD-L1 methylation restricts PD-L1/PD-1 interactions to control cancer immune surveillance. SCIENCE ADVANCES 2023; 9:eade4186. [PMID: 37235656 DOI: 10.1126/sciadv.ade4186] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 04/21/2023] [Indexed: 05/28/2023]
Abstract
Immune checkpoint inhibitors targeting programmed cell death protein 1 (PD-1) or programmed cell death 1 ligand 1 (PD-L1) have enabled some patients with cancer to experience durable, complete treatment responses; however, reliable anti-PD-(L)1 treatment response biomarkers are lacking. Our research found that PD-L1 K162 was methylated by SETD7 and demethylated by LSD2. Furthermore, PD-L1 K162 methylation controlled the PD-1/PD-L1 interaction and obviously enhanced the suppression of T cell activity controlling cancer immune surveillance. We demonstrated that PD-L1 hypermethylation was the key mechanism for anti-PD-L1 therapy resistance, investigated that PD-L1 K162 methylation was a negative predictive marker for anti-PD-1 treatment in patients with non-small cell lung cancer, and showed that the PD-L1 K162 methylation:PD-L1 ratio was a more accurate biomarker for predicting anti-PD-(L)1 therapy sensitivity. These findings provide insights into the regulation of the PD-1/PD-L1 pathway, identify a modification of this critical immune checkpoint, and highlight a predictive biomarker of the response to PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Changsheng Huang
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Yaqi Chen
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anyi Liu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Wu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Panjing Lv
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Da Song
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fuqing Hu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingqing Lan
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue Zheng
- Wuhan Blood Center, Wuhan 430030, China
| | - Xuelai Luo
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Keyi Jia
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jun Wang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Caicun Zou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Junbo Hu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guihua Wang
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
7
|
Donne R, Lujambio A. The liver cancer immune microenvironment: Therapeutic implications for hepatocellular carcinoma. Hepatology 2023; 77:1773-1796. [PMID: 35989535 PMCID: PMC9941399 DOI: 10.1002/hep.32740] [Citation(s) in RCA: 189] [Impact Index Per Article: 189.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 07/26/2022] [Accepted: 08/18/2022] [Indexed: 12/19/2022]
Abstract
The liver is the sixth most common site of primary cancer in humans and the fourth leading cause of cancer-related death in the world. Hepatocellular carcinoma (HCC) accounts for 90% of liver cancers. HCC is a prevalent disease with a progression that is modulated by the immune system. Half of the patients with HCC receive systemic therapies, traditionally sorafenib or lenvatinib, as a first-line therapy. In the last few years, immune-checkpoint inhibitors (ICIs) have revolutionized cancer therapy and have gained an increased interest in the treatment of HCC. In 2020, the combination of atezolizumab (anti-programmed death-ligand 1) and bevacizumab (anti-vascular endothelial growth factor) improved overall survival over sorafenib, resulting in Food and Drug Administration (FDA) approval as a first-line treatment for patients with advanced HCC. Despite these major advances, a better molecular and cellular characterization of the tumor microenvironment is still needed because it has a crucial role in the development and progression of HCC. Inflamed (hot) and noninflamed (cold) HCC tumors and genomic signatures have been associated with response to ICIs. However, there are no additional biomarkers to guide clinical decision-making. Other immune-targeting strategies, such as adoptive T-cell transfer, vaccination, and virotherapy, are currently under development. This review provides an overview on the HCC immune microenvironment, different cellular players, current available immunotherapies, and potential immunotherapy modalities.
Collapse
Affiliation(s)
- Romain Donne
- Department of Oncological Sciences , Icahn School of Medicine at Mount Sinai , New York , New York , USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai , Tisch Cancer Institute , New York , New York , USA
- Icahn School of Medicine at Mount Sinai , The Precision Immunology Institute , New York , New York , USA
| | - Amaia Lujambio
- Department of Oncological Sciences , Icahn School of Medicine at Mount Sinai , New York , New York , USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai , Tisch Cancer Institute , New York , New York , USA
- Icahn School of Medicine at Mount Sinai , The Precision Immunology Institute , New York , New York , USA
- Graduate School of Biomedical Sciences , Icahn School of Medicine at Mount Sinai , New York , New York , USA
| |
Collapse
|
8
|
Dong M, Zhang G, Meng J, Liu B, Jiang D, Liu F. MMP9-Associated Tumor Stem Cells, CCL1-Silenced Dendritic Cells, and Cytokine-Induced Killer Cells Have a Remarkable Therapeutic Efficacy for Acute Myeloid Leukemia by Activating T Cells. Stem Cells Int 2023; 2023:2490943. [PMID: 37200633 PMCID: PMC10188259 DOI: 10.1155/2023/2490943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 02/15/2023] [Accepted: 04/06/2023] [Indexed: 05/20/2023] Open
Abstract
Purpose Dendritic cells (DC) are specialized antigen-presenting cells, and cytokine-induced killer (CIK) cells have a specific killing activity to a variety of tumors. However, the underlining mechanism and function of DC-CIK cells in acute myeloid leukemia (AML) remain largely elusive. Methods Gene expression profiles of leukemia patients were obtained from TCGA, DC cell components were evaluated using the quanTIseq method, and cancer stem cell scores were estimated using machine learning methods. The transcriptomes were obtained in DC-CIK cells from normal and AML patients by high-throughput sequencing. Large differentially expressed mRNAs were verified by RT-qPCR assay, and MMP9 and CCL1 were selected for subsequent studies in vivo and in vitro experiments. Results Significant positive correlations were found with DC versus cancer stem cells (p = 0.008) and the expression of MMP9 versus cancer stem cells (p = 0.018). MMP9 and CCL1 were found to be highly expressed in DC-CIK cells from AML patients. DC-CIK cells with MMP9 and CCL1 knockout alone had little effect on leukemia cells, while knockdown of MMP9 and CCL1 in DC-CIK cells increased cytotoxicity, suppressed proliferation, and induced apoptosis of leukemia cells. In addition, we proved that MMP9- and CCL1-silenced DC-CIK cells significantly elevated the CD3+CD4+ and CD3+CD8+ cells and lowered the CD4+PD-1+ and CD8+PD-1+ T cells. Meanwhile, blockage of MMP9 and CCL1 in DC-CIK cells dramatically increased IL-2 and IFN-γ, increased CD107aþ (LAMP-1) and granzyme B (GZMB), and downregulated PD-1, CTLA4, TIM3, and LAG3 T cells from AML patients and AML model mice. Furthermore, activated T cells in DC-CIK cells knocking down MMP9 and CCL1 also prevented proliferation and accelerated apoptosis of AML cells. Conclusion Our findings demonstrated that blockage of MMP9 and CCL1 in DC-CIK cells could markedly enhance the therapeutic efficiency in AML via activating T cells.
Collapse
Affiliation(s)
- Min Dong
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570000, China
| | - Guozhen Zhang
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570000, China
| | - Jie Meng
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570000, China
| | - Biou Liu
- Department of Hematology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Duanfeng Jiang
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570000, China
| | - Feng Liu
- Department of Hematology, The Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| |
Collapse
|
9
|
Fu C, Ma T, Zhou L, Mi QS, Jiang A. Dendritic Cell-Based Vaccines Against Cancer: Challenges, Advances and Future Opportunities. Immunol Invest 2022; 51:2133-2158. [PMID: 35946383 DOI: 10.1080/08820139.2022.2109486] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
As the most potent professional antigen presenting cells, dendritic cells (DCs) have the ability to activate both naive CD4 and CD8 T cells. Recognized for their exceptional ability to cross-present exogenous antigens to prime naive antigen-specific CD8 T cells, DCs play a critical role in generating CD8 T cell immunity, as well as mediating CD8 T cell tolerance to tumor antigens. Despite the ability to potentiate host CD8 T cell-mediated anti-tumor immunity, current DC-based cancer vaccines have not yet achieved the promised success clinically with the exception of FDA-approved Provenge. Interestingly, recent studies have shown that type 1 conventional DCs (cDC1s) play a critical role in cross-priming tumor-specific CD8 T cells and determining the anti-tumor efficacy of cancer immunotherapies including immune checkpoint blockade (ICB). Together with promising clinical results in neoantigen-based cancer vaccines, there is a great need for DC-based vaccines to be further developed and refined either as monotherapies or in combination with other immunotherapies. In this review, we will present a brief review of DC development and function, discuss recent progress, and provide a perspective on future directions to realize the promising potential of DC-based cancer vaccines.
Collapse
Affiliation(s)
- Chunmei Fu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| | - Tianle Ma
- Department of Computer Science and Engineering, School of Engineering and Computer Science, Oakland University, Rochester, Michigan, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| | - Aimin Jiang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| |
Collapse
|
10
|
Wang D, Cui Q, Yang YJ, Liu AQ, Zhang G, Yu JC. Application of dendritic cells in tumor immunotherapy and progress in the mechanism of anti-tumor effect of Astragalus polysaccharide (APS) modulating dendritic cells: a review. Biomed Pharmacother 2022; 155:113541. [PMID: 36127221 DOI: 10.1016/j.biopha.2022.113541] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells (APCs) that are essential in mediating the body's natural and adaptive immune responses. The body can regulate the function of DCs in various ways to enhance their antitumor effects. In the tumour microenvironment (TME), antigen-specific T cell responses are initiated through DC processing and delivery of tumour-associated antigens (TAAs); conversely, tumour cells inhibit DC recruitment by releasing metabolites, cytokines and other regulatory TME and function. Different subpopulations of DCs exist in tumour tissues, and their functions vary. Insight into DC subgroups in TME allows assessment of the effectiveness of tumour immunotherapy. Astragalus polysaccharide (APS) is the main component of the Chinese herb Astragalus membranaceus. The study found that the antitumor effects of APS are closely related to DCs. APS can promote the expression of surface molecules CD80 and CD86, promote the maturation of DCs, and activate CTL to exert antitumor effects. We reviewed the application of DCs in tumor immunotherapy and the mechanism of modulation of DCs by Astragalus polysaccharide to provide new directions and strategies for tumor therapy and new drug development.
Collapse
Affiliation(s)
- Dong Wang
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Qian Cui
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Yan Jie Yang
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - A Qing Liu
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Guan Zhang
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Jian Chun Yu
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China.
| |
Collapse
|
11
|
Chen XY, Yung LYL, Tan PH, Bay BH. Harnessing the Immunogenic Potential of Gold Nanoparticle-Based Platforms as a Therapeutic Strategy in Breast Cancer Immunotherapy: A Mini Review. Front Immunol 2022; 13:865554. [PMID: 35432376 PMCID: PMC9008216 DOI: 10.3389/fimmu.2022.865554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/14/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer remains the most common malignancy among women worldwide. Although the implementation of mammography has dramatically increased the early detection rate, conventional treatments like chemotherapy, radiation therapy, and surgery, have significantly improved the prognosis for breast cancer patients. However, about a third of treated breast cancer patients are known to suffer from disease recurrences and progression to metastasis. Immunotherapy has recently gained traction due to its ability to establish long-term immune surveillance, and response for the prevention of disease recurrence and extension of patient survival. Current research findings have revealed that gold nanoparticles can enhance the safety and efficacy of cancer immunotherapy, through their unique intrinsic properties of good biocompatibility, durability, convenient surface modification, as well as enhanced permeability and retention effect. Gold nanoparticles are also able to induce innate immune responses through the process of immunogenic cell death, which can lead to the establishment of lasting adaptive immunity. As such gold nanoparticles are considered as good candidates for next generation immunotherapeutic strategies. This mini review gives an overview of gold nanoparticles and their potential applications in breast cancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Xiao-Yang Chen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Lin-Yue Lanry Yung
- Department of Biomolecular and Chemical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Puay Hoon Tan
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
12
|
Zhang T, Yang Y, Huang L, Liu Y, Chong G, Yin W, Dong H, Li Y, Li Y. Biomimetic and Materials-Potentiated Cell Engineering for Cancer Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14040734. [PMID: 35456568 PMCID: PMC9024915 DOI: 10.3390/pharmaceutics14040734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 02/01/2023] Open
Abstract
In cancer immunotherapy, immune cells are the main force for tumor eradication. However, they appear to be dysfunctional due to the taming of the tumor immunosuppressive microenvironment. Recently, many materials-engineered strategies are proposed to enhance the anti-tumor effect of immune cells. These strategies either utilize biomimetic materials, as building blocks to construct inanimate entities whose functions are similar to natural living cells, or engineer immune cells with functional materials, to potentiate their anti-tumor effects. In this review, we will summarize these advanced strategies in different cell types, as well as discussing the prospects of this field.
Collapse
Affiliation(s)
- Tingting Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Yushan Yang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Li Huang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Ying Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Gaowei Chong
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Weimin Yin
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| | - Haiqing Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
- Correspondence: (H.D.); (Y.L.); Tel.: +86-021-659-819-52 (H.D. & Y.L.)
| | - Yan Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
- Correspondence: (H.D.); (Y.L.); Tel.: +86-021-659-819-52 (H.D. & Y.L.)
| | - Yongyong Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China; (T.Z.); (Y.Y.); (L.H.); (Y.L.); (G.C.); (W.Y.); (Y.L.)
| |
Collapse
|
13
|
Anti-hepatoma Effect of DC2.4 Cells Transfected with Tumor-Associated Antigen Cdc25C In Vitro. Curr Med Sci 2022; 42:491-497. [PMID: 35292875 DOI: 10.1007/s11596-022-2556-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 10/25/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Cell division cyclin 25 homolog C (Cdc25C) is a tumor-associated antigen candidate gene, and this may be used as an effective target in cancer treatment. The present study aims to evaluate the lysis effect of cytotoxic T lymphocytes (CTLs) induced by dendritic cell line DC2.4 overexpressing Cdc25C, and the feasibility of Cdc25C as a component in hepatoma immunotherapy. METHODS The mouse Cdc25C gene was ligated into a lentiviral vector, and transfected into DC2.4 cells. The DC2.4 cell phenotype and cytokine secretion were determined by flow cytometry and ELISA, respectively. CD8+ T cells were sorted from the spleens of C57BL/6 mice using a magnetic bead sorting kit obtained from Miltenyi Biotech, Germany, and co-cultured with DC2.4 cells for one week as effector cells. Then, IL-2, granzyme B and perforin were detected in the CTL culture medium by ELISA. Next, time-resolved fluorescence immunoassay was used to detect the immune killing effect of Cdc25C-specific CTLs on target cells. Meanwhile, the effect of blocking MHC-I sites on target cells with a monoclonal anti-MHC-I antibody was evaluated. RESULTS The results revealed that Cdc25C could be stably overexpressed in DC2.4 cells by LV-Cdc25C infection. DC2.4 cells transfected with LV-Cdc25C secreted more IL-6, IL-12, TNF-α and IFN-γ, and had higher expression levels of CD40, CD86, CCR7 and MHC-II than unaltered DC2.4 cells. The elevated Cdc25C in dendritic cells also further increased the secretion of IL-2, granzyme B and perforin to elicit Cdc25C-specific CTLs, and induced the higher cytotoxicity in Hepa1-6 cell lines (P<0.05), but this had no effect on the target cells when MHC-I monoclonal antibodies were blocked. CONCLUSION DC2.4 cells transfected with LV-Cdc25C can induce specific CTLs, and result in a strong cellular immune response. The dendritic cells that overexpress Cdc25C may be useful for hepatoma immunotherapy.
Collapse
|
14
|
Liu K, Liao Y, Zhou Z, Zhang L, Jiang Y, Lu H, Xu T, Yang D, Gao Q, Li Z, Tan S, Cao W, Chen F, Li G. Photothermal-triggered immunogenic nanotherapeutics for optimizing osteosarcoma therapy by synergizing innate and adaptive immunity. Biomaterials 2022; 282:121383. [DOI: 10.1016/j.biomaterials.2022.121383] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 12/20/2022]
|
15
|
Uetz-von Allmen E, Samson GPB, Purvanov V, Maeda T, Legler DF. CAL-1 as Cellular Model System to Study CCR7-Guided Human Dendritic Cell Migration. Front Immunol 2021; 12:702453. [PMID: 34603281 PMCID: PMC8482423 DOI: 10.3389/fimmu.2021.702453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/31/2021] [Indexed: 11/23/2022] Open
Abstract
Dendritic cells (DCs) are potent and versatile professional antigen-presenting cells and central for the induction of adaptive immunity. The ability to migrate and transport peripherally acquired antigens to draining lymph nodes for subsequent cognate T cell priming is a key feature of DCs. Consequently, DC-based immunotherapies are used to elicit tumor-antigen specific T cell responses in cancer patients. Understanding chemokine-guided DC migration is critical to explore DCs as cellular vaccines for immunotherapeutic approaches. Currently, research is hampered by the lack of appropriate human cellular model systems to effectively study spatio-temporal signaling and CCR7-driven migration of human DCs. Here, we report that the previously established human neoplastic cell line CAL-1 expresses the human DC surface antigens CD11c and HLA-DR together with co-stimulatory molecules. Importantly, if exposed for three days to GM-CSF, CAL-1 cells induce the endogenous expression of the chemokine receptor CCR7 upon encountering the clinically approved TLR7/8 agonist Resiquimod R848 and readily migrate along chemokine gradients. Further, we demonstrate that CAL-1 cells can be genetically modified to express fluorescent (GFP)-tagged reporter proteins to study and visualize signaling or can be gene-edited using CRISPR/Cas9. Hence, we herein present the human CAL-1 cell line as versatile and valuable cellular model system to effectively study human DC migration and signaling.
Collapse
Affiliation(s)
- Edith Uetz-von Allmen
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
| | - Guerric P B Samson
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland.,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Vladimir Purvanov
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
| | - Takahiro Maeda
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland.,Theodor Kocher Institute, University of Bern, Bern, Switzerland.,Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
16
|
Zhou L, Liu H, Liu K, Wei S. Gold Compounds and the Anticancer Immune Response. Front Pharmacol 2021; 12:739481. [PMID: 34588987 PMCID: PMC8473785 DOI: 10.3389/fphar.2021.739481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022] Open
Abstract
Gold compounds are not only well-explored for cytotoxic effects on tumors, but are also known to interact with the cancer immune system. The immune system deploys innate and adaptive mechanisms to protect against pathogens and prevent malignant transformation. The combined action of gold compounds with the activated immune system has shown promising results in cancer therapy through in vivo and in vitro experiments. Gold compounds are known to induce innate immune responses; however, these responses may contribute to adaptive immune responses. Gold compounds play the role of a major hapten that acts synergistically in innate immunity. Gold compounds support cancer cell antigenicity and promote anti-tumor immune response by inducing the release of CRT, ATP, HMGB1, HSP, and NKG2D to enhance immunogenicity. Gold compounds affect various immune cells (including suppressor regulatory T cells), inhibit myeloid derived suppressor cells, and enhance the function and number of dendritic cells. Gold nanoparticles (AuNPs) have potential for improving the effect of immunotherapy and reducing the toxicity and side effects of the treatment process. Thus, AuNPs provide an ideal opportunity for exploring the combination of anticancer gold compounds and immunotherapeutic interventions.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Kui Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Zhang L, Xia H, Xia K, Liu X, Zhang X, Dai J, Zeng Z, Jia Y. Selenium Regulation of the Immune Function of Dendritic Cells in Mice Through the ERK, Akt and RhoA/ROCK Pathways. Biol Trace Elem Res 2021; 199:3360-3370. [PMID: 33107016 DOI: 10.1007/s12011-020-02449-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/18/2020] [Indexed: 10/23/2022]
Abstract
Selenium levels can regulate the function of T cells, macrophages, B cells, natural killer cells and other immune cells. However, the effect of selenium on the immune function of dendritic cells (DCs) isolated from selenium-supplemented mice is unknown. In this study, C57BL/6J mice were randomly divided into three groups and fed diets containing low (0.08 ppm), medium (0.25 ppm) or high (1 ppm) selenium levels for 8 weeks. Immature (imDCs) and mature (mDCs) dendritic cells were then isolated from the bone marrow. Next, the migration, phagocytic capacity and mixed lymphocyte reaction (MLR) for imDCs and mDCs were detected by transwell and flow cytometry. The levels of C-C chemokine receptor type 7 (CCR7), major histocompatibility complex II (MHCII) and reactive oxygen species (ROS) were assayed by flow cytometry. F-actin and superoxide dismutase (SOD) activity was detected by fluorescence microscopy and SOD assay kit, respectively. In addition, the extracellular signal-regulated kinase (ERK), Akt, Ras homolog gene family member A/Rho-associated protein kinase (RhoA/ROCK) signalling, selenoprotein K (SELENOK) and glutathione peroxidase 1 (GPX1) levels were measured by western blot analysis. The results indicated that selenium deficiency enhanced the migration of imDCs by ROS and SELENOK-mediated ERK, Akt and RhoA/ROCK pathways but impaired the antigen uptake of imDCs. Although a high selenium level inhibited the migration of imDCs, it had no effect on phagocytic capacity. For mDCs, low selenium levels impaired free migration, and high levels inhibited the chemotactic migration involved in F-actin and CCR7, respectively. Low and high selenium levels impaired the MLR by inhibiting MHCII surface localisation, which might be related to ROS- and SELENOK-mediated ERK, Akt and RhoA/ROCK signalling pathways. In summary, selenium may regulate the immune function of mouse DCs through the ROS- and SELENOK-mediated ERK, Akt and RhoA/ROCK signalling.
Collapse
Affiliation(s)
- Liangliang Zhang
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Huan Xia
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Kaide Xia
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xianmei Liu
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xin Zhang
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jie Dai
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhu Zeng
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Basic Medical Science, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yi Jia
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China.
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
18
|
Feng H, Yang X, Zhang L, Liu Q, Feng Y, Wu D, Liu Y, Yang J. Mannose-Modified Chitosan Poly(lactic- co-glycolic acid) Microspheres Act as a Mannose Receptor-Mediated Delivery System Enhancing the Immune Response. Polymers (Basel) 2021; 13:polym13132208. [PMID: 34279352 PMCID: PMC8271610 DOI: 10.3390/polym13132208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 12/16/2022] Open
Abstract
The mannose receptor (MAN-R)-targeted delivery system is commonly used to deliver antigens to macrophages or immature dendritic cells (DCs) to promote the efficiency of antigen presentation. To maximize the enhancement effects of chitosan (CS) and induce an efficient humoral and cellular immune response against an antigen, we encapsulated ovalbumin (OVA) in poly(lactic-co-glycolic acid) (PLGA) microspheres (MPs) and conjugated it with MAN-modified CS to obtain MAN-R-targeting nano-MPs (MAN-CS-OVA-PLGA-MPs). The physicochemical properties, drug loading rate, and immunomodulation activity of MAN-CS-OVA-PLGA-MPs were evaluated. In vitro, MAN-CS-OVA-PLGA-MPs (80 μg mL−1) could enhance the proliferation of DCs and increase their phagocytic efficiency. In vivo, MAN-CS-OVA-PLGA-MPs significantly increased the ratio of CD3+CD4+/CD3+CD8+ T cells, increased CD80+, CD86+, and MHC II expression in DCs, and improved OVA-specific IgG, IgG1, IgG2a, and IgG2b antibodies. Moreover, MAN-CS-OVA-PLGA-MPs promoted cytokine (IFN-γ, IL-4, and IL-6) production in mice. Taken together, our results show that MAN-CS-OVA-PLGA-MPs may act by activating the T cells to initiate an immune response by promoting the maturation of dendritic cells and improving their antigen presentation efficiency. The current study provides a basis for the use of MAN-CS-OVA-PLGA-MPs as an antigen and adjuvant delivery system targeting the MAN-R on the surface of macrophages and dendritic cells.
Collapse
Affiliation(s)
- Haibo Feng
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (X.Y.); (L.Z.); (Q.L.); (Y.F.); (D.W.)
- Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu 610041, China
- Correspondence: ; Tel./Fax: +86-28-85522310
| | - Xiaonong Yang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (X.Y.); (L.Z.); (Q.L.); (Y.F.); (D.W.)
- Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu 610041, China
| | - Linzi Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (X.Y.); (L.Z.); (Q.L.); (Y.F.); (D.W.)
- Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu 610041, China
| | - Qianqian Liu
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (X.Y.); (L.Z.); (Q.L.); (Y.F.); (D.W.)
- Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu 610041, China
| | - Yangyang Feng
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (X.Y.); (L.Z.); (Q.L.); (Y.F.); (D.W.)
- Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu 610041, China
| | - Daiyan Wu
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (X.Y.); (L.Z.); (Q.L.); (Y.F.); (D.W.)
- Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Chengdu 610041, China
| | - Yunjie Liu
- Department of Veterinary Medicine, Southwest University, Rongchang 402460, China; (Y.L.); (J.Y.)
| | - Jie Yang
- Department of Veterinary Medicine, Southwest University, Rongchang 402460, China; (Y.L.); (J.Y.)
| |
Collapse
|
19
|
Teng CF, Wang T, Shih FY, Shyu WC, Jeng LB. Therapeutic efficacy of dendritic cell vaccine combined with programmed death 1 inhibitor for hepatocellular carcinoma. J Gastroenterol Hepatol 2021; 36:1988-1996. [PMID: 33462840 DOI: 10.1111/jgh.15398] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/15/2020] [Accepted: 01/01/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Hepatocellular carcinoma (HCC) remains a serious cause of cancer-related deaths worldwide. Developing new therapeutic strategies is urgently needed to improve the outcomes of HCC patients. Dendritic cell (DC)-based vaccines and programmed death 1 (PD-1) immune checkpoint inhibitors have been regarded as potential immunotherapeutics for HCC. However, the therapeutic efficacy of combining these two treatments for HCC remains to be evaluated. METHODS In this study, DCs were derived from mouse bone marrow and pulsed with mouse HCC cell lysates to generate a DC vaccine. A monoclonal antibody that blocks the interaction of mouse PD-1 with its ligands was used as a PD-1 inhibitor. An orthotopic HCC mouse model was established to assess the effect of a DC vaccine in combination with a PD-1 inhibitor on overall survival and tumor volume. RESULTS Compared with the untreated control, single treatment with a DC vaccine or PD-1 inhibitor prolonged the overall survival and reduced the tumor volume of HCC mice. Further, compared with the single treatment with the DC vaccine or the PD-1 inhibitor, a combination treatment using both agents elicited a higher cytotoxicity of T cells against HCC cells and resulted in a better overall survival, smaller tumor volume, and greater tumor cell apoptosis in HCC mice. CONCLUSIONS Our results suggest that a combination treatment with DC vaccine and PD-1 inhibitor may be a promising therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Chiao-Fang Teng
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan.,Research Center for Cancer Biology, China Medical University, Taichung, Taiwan
| | - Ting Wang
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| | - Fu-Ying Shih
- Ph.D. Program for Biotech Pharmaceutical Industry, School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Woei-Cherng Shyu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Occupational Therapy, Asia University, Taichung, Taiwan.,Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
20
|
Characterization of a Tumor-Microenvironment-Relevant Gene Set Based on Tumor Severity in Colon Cancer and Evaluation of Its Potential for Dihydroartemisinin Targeting. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:4812068. [PMID: 34239578 PMCID: PMC8233087 DOI: 10.1155/2021/4812068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/09/2021] [Indexed: 01/12/2023]
Abstract
Colon cancer (COAD) is a leading cause of cancer mortality in the world. Most patients with COAD die as a result of cancer cell metastasis. However, the mechanisms underlying the metastatic phenotype of COAD remain unclear. Instead, particular features of the tumor microenvironment (TME) could predict adverse outcomes including metastasis in patients with COAD, and the role of TME in governing COAD progression is undeniable. Therefore, exploring the role of TME in COAD may help us better understand the molecular mechanisms behind COAD progression which may improve clinical outcomes and quality of patients. Here, we identified a Specific TME Regulatory Network including AEBP1, BGN, POST, and FAP (STMERN) that is highly involved in clinical outcomes of patients with COAD. Comprehensive in silico analysis of our study revealed that the STMERN is highly correlated with the severity of COAD. Meanwhile, our results reveal that the STMERN might be associated with immune infiltration in COAD. Importantly, we show that dihydroartemisinin (DHA) potentially interacts with the STMERN. We suggest that DHA might contribute to immune infiltration through regulating the STMERN in COAD. Taken together, our data provide a set of biomarkers of progression and poor prognosis in COAD. These findings could have potential prognostic and therapeutic implications in the progression of COAD.
Collapse
|
21
|
Preclinical models and technologies to advance nanovaccine development. Adv Drug Deliv Rev 2021; 172:148-182. [PMID: 33711401 DOI: 10.1016/j.addr.2021.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
The remarkable success of targeted immunotherapies is revolutionizing cancer treatment. However, tumor heterogeneity and low immunogenicity, in addition to several tumor-associated immunosuppression mechanisms are among the major factors that have precluded the success of cancer vaccines as targeted cancer immunotherapies. The exciting outcomes obtained in patients upon the injection of tumor-specific antigens and adjuvants intratumorally, reinvigorated interest in the use of nanotechnology to foster the delivery of vaccines to address cancer unmet needs. Thus, bridging nano-based vaccine platform development and predicted clinical outcomes the selection of the proper preclinical model will be fundamental. Preclinical models have revealed promising outcomes for cancer vaccines. However, only few cases were associated with clinical responses. This review addresses the major challenges related to the translation of cancer nano-based vaccines to the clinic, discussing the requirements for ex vivo and in vivo models of cancer to ensure the translation of preclinical success to patients.
Collapse
|
22
|
Saichi M, Ladjemi MZ, Korniotis S, Rousseau C, Ait Hamou Z, Massenet-Regad L, Amblard E, Noel F, Marie Y, Bouteiller D, Medvedovic J, Pène F, Soumelis V. Single-cell RNA sequencing of blood antigen-presenting cells in severe COVID-19 reveals multi-process defects in antiviral immunity. Nat Cell Biol 2021; 23:538-551. [PMID: 33972731 DOI: 10.1038/s41556-021-00681-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 04/06/2021] [Indexed: 02/03/2023]
Abstract
COVID-19 can lead to life-threatening respiratory failure, with increased inflammatory mediators and viral load. Here, we perform single-cell RNA-sequencing to establish a high-resolution map of blood antigen-presenting cells (APCs) in 15 patients with moderate or severe COVID-19 pneumonia, at day 1 and day 4 post admission to intensive care unit or pulmonology department, as well as in 4 healthy donors. We generated a unique dataset of 81,643 APCs, including monocytes and rare dendritic cell (DC) subsets. We uncovered multi-process defects in antiviral immune defence in specific APCs from patients with severe disease: (1) increased pro-apoptotic pathways in plasmacytoid DCs (pDCs, key effectors of antiviral immunity), (2) a decrease of the innate sensors TLR9 and DHX36 in pDCs and CLEC9a+ DCs, respectively, (3) downregulation of antiviral interferon-stimulated genes in monocyte subsets and (4) a decrease of major histocompatibility complex (MHC) class II-related genes and MHC class II transactivator activity in cDC1c+ DCs, suggesting viral inhibition of antigen presentation. These novel mechanisms may explain patient aggravation and suggest strategies to restore the defective immune defence.
Collapse
Affiliation(s)
| | - Maha Zohra Ladjemi
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, Paris, France
- Service de Médecine Intensive & Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris. Centre & Université de Paris, Paris, France
| | | | - Christophe Rousseau
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, Paris, France
| | - Zakaria Ait Hamou
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, Paris, France
- Service de Médecine Intensive & Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris. Centre & Université de Paris, Paris, France
| | - Lucile Massenet-Regad
- Université de Paris, INSERM U976, Paris, France
- Université Paris-Saclay, Saint-Aubin, France
| | - Elise Amblard
- Université de Paris, INSERM U976, Paris, France
- Université de Paris, Centre de Recherches Interdisciplinaires, Paris, France
| | | | - Yannick Marie
- Institut du Cerveau (ICM), Plateforme de Génotypage Séquençage, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie, Paris, France
| | - Delphine Bouteiller
- Institut du Cerveau (ICM), Plateforme de Génotypage Séquençage, Paris, France
| | | | - Frédéric Pène
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, Paris, France
- Service de Médecine Intensive & Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris. Centre & Université de Paris, Paris, France
| | - Vassili Soumelis
- Université de Paris, INSERM U976, Paris, France.
- AP-HP, Hôpital Saint-Louis, Laboratoire d'Immunologie-Histocompatibilité, Paris, France.
| |
Collapse
|
23
|
Wang W, Li H, Zhang L, Jiang W, Shen L, Fan G. Clinical applications of monitoring immune status with 90 immune cell subsets in human whole blood by 10-color flow cytometry. Int J Lab Hematol 2021; 43:1132-1144. [PMID: 33870648 DOI: 10.1111/ijlh.13541] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/19/2021] [Accepted: 03/26/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The immune system may involve and predict the different prognosis and therapy consequences. So, it's important to monitor and evaluate the immune status before and after treatments. METHODS Flow cytometry is the best technology to perform immune monitoring, because it can detect immune cells using small amount of sample in a short time. The whole blood is the ideal sample for immune status monitoring, since it includes almost all the immune cells and it's relatively easy to obtain and less invasive than bone marrow or lymph node. RESULTS Here we developed and validated a 10-color panel with only four tubes containing 29 antibodies to monitor 90 immune cell subsets in 2 ml whole blood samples. The major immune cell populations detected by our panel included T cell subsets (CD3+ total T, Th, Tc, Treg, CD8hi , CD8low , αβTCR, γδTCR, naïve, and memory T), T cell activation markers (CD25, CD69, and HLA-DR) and one immune checkpoint PD1, B cell subsets (B1, switched memory, non-switched, naïve B, and CD27- IgD- B cells), neutrophils, basophils, four monocytic cell subsets, dendritic cells (pDCs and mDCs), and four NK cell subsets. These panels of antibodies had been applied to monitor immune status (percentage and absolute number) in total 303 cases with various diseases, such as leukemia (AML, CML, MM, and ALL), lymphoma (B cells and NK/T cells), cancers (colon, lung, prostate, and breast), immune deficiencies, and autoimmune diseases. CONCLUSION We provided proof of feasibility for clinical monitoring immune status and guiding immunotherapy by multicolor flow cytometry testing.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Clinical laboratory, Xinhua hospital, Shanghai Jiaotong University of Medicine School, Shanghai, China
| | - Haibo Li
- Department of Pathology, Oregon Health and Science University, Portland, OR, USA
| | - Lihua Zhang
- Department of Clinical laboratory, Xinhua hospital, Shanghai Jiaotong University of Medicine School, Shanghai, China
| | - Wenli Jiang
- Department of Clinical laboratory, Xinhua hospital, Shanghai Jiaotong University of Medicine School, Shanghai, China
| | - Lisong Shen
- Department of Clinical laboratory, Xinhua hospital, Shanghai Jiaotong University of Medicine School, Shanghai, China
| | - Guang Fan
- Department of Pathology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
24
|
Thakur N, Thakur S, Chatterjee S, Das J, Sil PC. Nanoparticles as Smart Carriers for Enhanced Cancer Immunotherapy. Front Chem 2020; 8:597806. [PMID: 33409265 PMCID: PMC7779678 DOI: 10.3389/fchem.2020.597806] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy for the treatment of many forms of cancer by stimulating body's own immune system. This therapy not only eradicates tumor cells by inducing strong anti-tumor immune response but also prevent their recurrence. The clinical cancer immunotherapy faces some insurmountable challenges including high immune-mediated toxicity, lack of effective and targeted delivery of cancer antigens to immune cells and off-target side effects. However, nanotechnology offers some solutions to overcome those limitations, and thus can potentiate the efficacy of immunotherapy. This review focuses on the advancement of nanoparticle-mediated delivery of immunostimulating agents for efficient cancer immunotherapy. Here we have outlined the use of the immunostimulatory nanoparticles as a smart carrier for effective delivery of cancer antigens and adjuvants, type of interactions between nanoparticles and the antigen/adjuvant as well as the factors controlling the interaction between nanoparticles and the receptors on antigen presenting cells. Besides, the role of nanoparticles in targeting/activating immune cells and modulating the immunosuppressive tumor microenvironment has also been discussed extensively. Finally, we have summarized some theranostic applications of the immunomodulatory nanomaterials in treating cancers based on the earlier published reports.
Collapse
Affiliation(s)
- Neelam Thakur
- Himalayan Centre for Excellence in Nanotechnology, Shoolini University, Solan, India
- School of Advanced Chemical Sciences, Faculty of Basic Sciences, Shoolini University, Solan, India
| | - Saloni Thakur
- Himalayan Centre for Excellence in Nanotechnology, Shoolini University, Solan, India
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, India
| | | | - Joydeep Das
- Himalayan Centre for Excellence in Nanotechnology, Shoolini University, Solan, India
- School of Advanced Chemical Sciences, Faculty of Basic Sciences, Shoolini University, Solan, India
| | - Parames C. Sil
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
25
|
Abstract
Over the last decade, there has been a considerable progress in the development of cell therapy products for the treatment of liver diseases. The quest to generate well-defined homogenous cell populations with defined mechanism(s) of action has enabled the progression from use of autologous bone marrow stem cells comprising of heterogeneous cell populations to allogeneic cell types such as monocyte-derived macrophages, regulatory T cells, mesenchymal stromal cells, macrophages, etc. There is growing evidence regarding the multiple molecular mechanisms pivotal to various therapeutic effects and hence, careful selection of cell therapy product for the desired putative effects is crucial. In this review, we have presented an overview of the cell therapies that have been developed thus far, with preclinical and clinical evidence for their use in liver disease. Limitations associated with these therapies have also been discussed. Despite the advances made, there remain multiple challenges to overcome before cell therapies can be considered as viable treatment options, and these include larger scale clinical trials, scalable production of cells according to good manufacturing practice standards, pathways for delivery of cell therapy within hospital environments, and costs associated with the production.
Collapse
Affiliation(s)
- Sheeba Khan
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Reenam S Khan
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Philip N Newsome
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
26
|
Yu L, Feng R, Zhu L, Hao Q, Chu J, Gu Y, Luo Y, Zhang Z, Chen G, Chen H. Promoting the activation of T cells with glycopolymer-modified dendritic cells by enhancing cell interactions. SCIENCE ADVANCES 2020; 6:eabb6595. [PMID: 33219021 PMCID: PMC7679162 DOI: 10.1126/sciadv.abb6595] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/08/2020] [Indexed: 05/12/2023]
Abstract
Dendritic cell (DC) modification to enhance antigen presentation is a valuable strategy in cancer immune therapy. Other than focusing on regulating interactions between DC and antigens, we intend to promote cell interactions between DC and T cell by cell surface engineering. T cell activation is greatly improved and generates higher tumor toxicity with the aid of the synthetic glycopolymer modified on the DC surface, although the glycopolymer alone shows no effect. The great promotion of DC-T cell attraction is revealed by cell image tracking in terms of both frequency and duration of contacts. Our findings provide a new method of T cell activation by these engineered "sweet DCs." This strategy is beneficial for developing more efficient DC-based vaccines.
Collapse
Affiliation(s)
- Liyin Yu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Ruyan Feng
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Lijuan Zhu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Qing Hao
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Jiacheng Chu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Yan Gu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Yan Luo
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Zexin Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China
| | - Gaojian Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China.
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, P. R. China
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, P. R. China.
| |
Collapse
|
27
|
Teng CF, Wang T, Wu TH, Lin JH, Shih FY, Shyu WC, Jeng LB. Combination therapy with dendritic cell vaccine and programmed death ligand 1 immune checkpoint inhibitor for hepatocellular carcinoma in an orthotopic mouse model. Ther Adv Med Oncol 2020; 12:1758835920922034. [PMID: 32565925 PMCID: PMC7288802 DOI: 10.1177/1758835920922034] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/03/2020] [Indexed: 02/06/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is among the most common and lethal human cancers worldwide. Despite remarkable advances in treatment, high mortality in HCC patients remains a big challenge. To develop novel therapeutic strategies for HCC is thus urgently needed to improve patient survival. Dendritic cells (DC)-based vaccines can induce tumor-specific immunity and have emerged as a promising approach for treating HCC patients; however, its effectiveness needs to be improved. Recently, blockade of programmed death ligand 1 (PD-L1) immune checkpoint pathway has been shown to enhance anti-tumor immune responses and exhibited great potential in HCC therapy. Methods In this study, we generated DC vaccine by pulsing the C57BL/6J mouse bone marrow-derived DC with mouse hepatoma Hep-55.1C cell lysate. We developed a therapeutic strategy combining DC vaccine and PD-L1 inhibitor for HCC and evaluated its efficacy in an orthotopic HCC mouse model in which Hep-55.1C cells were directly injected into left liver lobe of C57BL/6J mouse. Results Compared with a control group of mice, groups of mice treated with DC vaccine or PD-L1 inhibitor had significantly improved overall survival, reduced tumor volume, and increased tumor cell apoptosis. Remarkably, combination treatment with DC vaccine and PD-L1 inhibitor led to considerably longer overall survival, smaller tumor volume, and higher tumor cell apoptosis of mice than either treatment alone in a dose-dependent manner through inducing a stronger anti-tumor cytotoxic T cell response. Conclusion Our data suggested that combination therapy with DC vaccine and PD-L1 inhibitor might have great promise as a novel treatment strategy for HCC.
Collapse
Affiliation(s)
- Chiao-Fang Teng
- Graduate Institute of Biomedical Sciences, China Medical University, No.91, Hsueh-Shih Rd., Northern Dist., Taichung City 404Organ Transplantation Center, China Medical University Hospital, Taichung Research Center for Cancer Biology, China Medical University, Taichung
| | - Ting Wang
- Organ Transplantation Center, China Medical University Hospital, Taichung
| | - Tzu-Hua Wu
- Organ Transplantation Center, China Medical University Hospital, Taichung
| | - Jia-Hui Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung
| | - Fu-Ying Shih
- Program for Biotech Pharmaceutical Industry, School of Pharmacy, China Medical University, Taichung
| | - Woei-Cherng Shyu
- Graduate Institute of Biomedical Sciences, China Medical University, No.91, Hsueh-Shih Rd., Northern Dist., Taichung City 404 Department of Occupational Therapy, Asia University, Taichung Department of Neurology, China Medical University Hospital, Taichung Translational Medicine Research Center, China Medical University Hospital, Taichung
| | - Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, No.2, Yude Rd., Northern Dist., Taichung City 404
| |
Collapse
|
28
|
Harding SD, Faccenda E, Southan C, Pawson AJ, Maffia P, Alexander SPH, Davenport AP, Fabbro D, Levi‐Schaffer F, Spedding M, Davies JA. The IUPHAR Guide to Immunopharmacology: connecting immunology and pharmacology. Immunology 2020; 160:10-23. [PMID: 32020584 PMCID: PMC7160657 DOI: 10.1111/imm.13175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/17/2020] [Accepted: 01/27/2020] [Indexed: 12/19/2022] Open
Abstract
Given the critical role that the immune system plays in a multitude of diseases, having a clear understanding of the pharmacology of the immune system is crucial to new drug discovery and development. Here we describe the International Union of Basic and Clinical Pharmacology (IUPHAR) Guide to Immunopharmacology (GtoImmuPdb), which connects expert-curated pharmacology with key immunological concepts and aims to put pharmacological data into the hands of immunologists. In the pursuit of new therapeutics, pharmacological databases are a vital resource to researchers through providing accurate information on the fundamental science underlying drug action. This extension to the existing IUPHAR/British Pharmacological Society Guide to Pharmacology supports research into the development of drugs targeted at modulating immune, inflammatory or infectious components of disease. To provide a deeper context for how the resource can support research we show data in GtoImmuPdb relating to a case study on the targeting of vascular inflammation.
Collapse
Affiliation(s)
- Simon D. Harding
- Deanery of Biomedical SciencesUniversity of EdinburghEdinburghUK
| | - Elena Faccenda
- Deanery of Biomedical SciencesUniversity of EdinburghEdinburghUK
| | - Christopher Southan
- Deanery of Biomedical SciencesUniversity of EdinburghEdinburghUK
- Present address:
TW2Informatics LtdGöteborg42166Sweden
| | - Adam J. Pawson
- Deanery of Biomedical SciencesUniversity of EdinburghEdinburghUK
| | - Pasquale Maffia
- Centre for ImmunobiologyInstitute of Infection, Immunity and InflammationCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
- Institute of Cardiovascular and Medical SciencesCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | | | | | - Doriano Fabbro
- Cellestia Biotech SABaselSwitzerland
- TargImmune Therapeutics AGBaselSwitzerland
| | | | | | - Jamie A. Davies
- Deanery of Biomedical SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
29
|
Bouzid R, Peppelenbosch M, Buschow SI. Opportunities for Conventional and in Situ Cancer Vaccine Strategies and Combination with Immunotherapy for Gastrointestinal Cancers, A Review. Cancers (Basel) 2020; 12:cancers12051121. [PMID: 32365838 PMCID: PMC7281593 DOI: 10.3390/cancers12051121] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
Survival of gastrointestinal cancer remains dismal, especially for metastasized disease. For various cancers, especially melanoma and lung cancer, immunotherapy has been proven to confer survival benefits, but results for gastrointestinal cancer have been disappointing. Hence, there is substantial interest in exploring the usefulness of adaptive immune system education with respect to anti-cancer responses though vaccination. Encouragingly, even fairly non-specific approaches to vaccination and immune system stimulation, involving for instance influenza vaccines, have shown promising results, eliciting hopes that selection of specific antigens for vaccination may prove useful for at least a subset of gastrointestinal cancers. It is widely recognized that immune recognition and initiation of responses are hampered by a lack of T cell help, or by suppressive cancer-associated factors. In this review we will discuss the hurdles that limit efficacy of conventional cancer therapeutic vaccination methods (e.g., peptide vaccines, dendritic cell vaccination). In addition, we will outline other forms of treatment (e.g., radiotherapy, chemotherapy, oncolytic viruses) that also cause the release of antigens through immunogenic tumor cell death and can thus be considered unconventional vaccination methods (i.e., in situ vaccination). Finally, we focus on the potential additive value that vaccination strategies may have for improving the effect immunotherapy. Overall, a picture will emerge that although the field has made substantial progress, successful immunotherapy through the combination with cancer antigen vaccination, including that for gastrointestinal cancers, is still in its infancy, prompting further intensification of the research effort in this respect.
Collapse
|
30
|
Zuo B, Qi H, Lu Z, Chen L, Sun B, Yang R, Zhang Y, Liu Z, Gao X, You A, Wu L, Jing R, Zhou Q, Yin H. Alarmin-painted exosomes elicit persistent antitumor immunity in large established tumors in mice. Nat Commun 2020; 11:1790. [PMID: 32286296 PMCID: PMC7156382 DOI: 10.1038/s41467-020-15569-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 03/17/2020] [Indexed: 11/09/2022] Open
Abstract
Treating large established tumors is challenging for dendritic cell (DC)-based immunotherapy. DC activation with tumor cell-derived exosomes (TEXs) carrying multiple tumor-associated antigen can enhance tumor recognition. Adding a potent adjuvant, high mobility group nucleosome-binding protein 1 (HMGN1), boosts DCs’ ability to activate T cells and improves vaccine efficiency. Here, we demonstrate that TEXs painted with the functional domain of HMGN1 (TEX-N1ND) via an exosomal anchor peptide potentiates DC immunogenicity. TEX-N1ND pulsed DCs (DCTEX-N1ND) elicit long-lasting antitumor immunity and tumor suppression in different syngeneic mouse models with large tumor burdens, most notably large, poorly immunogenic orthotopic hepatocellular carcinoma (HCC). DCTEX-N1ND show increased homing to lymphoid tissues and contribute to augmented memory T cells. Importantly, N1ND-painted serum exosomes from cancer patients also promote DC activation. Our study demonstrates the potency of TEX-N1ND to strengthen DC immunogenicity and to suppress large established tumors, and thus provides an avenue to improve DC-based immunotherapy. The use of tumour exosome-activated dendritic cell (DC) immunotherapy shows promise for the treatment of large established tumours. Here, the authors generate alarmin HMGN1-attached tumour exosomes which significantly improve therapy efficacy by boosting DC activation in several preclinical mouse models.
Collapse
Affiliation(s)
- Bingfeng Zuo
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases & Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Han Qi
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases & Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Zhen Lu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases & Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Lu Chen
- Department of Hepatobiliary, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Bo Sun
- Department of The Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Rong Yang
- Department of Nanomedicine & Biopharmaceuticals, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, China
| | - Yang Zhang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases & Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Zhili Liu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases & Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Xianjun Gao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases & Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Abin You
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases & Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Li Wu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases & Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Renwei Jing
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases & Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Qibing Zhou
- Department of Nanomedicine & Biopharmaceuticals, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, China
| | - HaiFang Yin
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) & Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases & Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China.
| |
Collapse
|
31
|
Danilova A, Misyurin V, Novik A, Girdyuk D, Avdonkina N, Nekhaeva T, Emelyanova N, Pipia N, Misyurin A, Baldueva I. Cancer/testis antigens expression during cultivation of melanoma and soft tissue sarcoma cells. Clin Sarcoma Res 2020; 10:3. [PMID: 32042403 PMCID: PMC6998350 DOI: 10.1186/s13569-020-0125-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/22/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Autologous dendritic cells (DC) loaded with tumor-associated antigens (TAAs) are a promising approach for anticancer immunotherapy. Polyantigen lysates appear to be an excellent source of TAAs for loading onto the patient's dendritic cells. Cancer/testis antigens (CTA) are expressed by a wide range of tumors, but are minimally expressed on normal tissues, and could serve as a universal target for immunotherapy. However, CTA expression levels can vary significantly in patients with the same tumor type. We proposed that patients who do not respond to DC-based therapy may have distinct features of the CTA expression profile on tumor cells. PATIENTS AND METHODS We compared the gene expression of the principal families CTA in 22 melanoma and 27 soft tissue and bone sarcomas cell lines (STBS), received from patients and used for DC vaccine preparation. RESULTS The majority (47 of 49, 95.9%) cell lines showed CTA gene activity. The incidence of gene expression of GAGE, NYESO1, MAGEA1, PRAME's was significantly different (adj. p < 0.05) between melanoma and sarcoma cell lines. The expression of the SCP1 gene was detected neither in melanoma cells nor in the STBS cells. Clustering by the gene expression profile revealed four different expression patterns. We found three main patterns types: hyperexpression of multiple CTA, hyperexpression of one CTA with almost no expression of others, and no expression of CTA. All clusters types exist in melanoma and sarcoma cell lines. We observed dependence of killing efficacy from the PRAME (rho = 0.940, adj. p < 0.01) expression during real-time monitoring with the xCELLigence system of the interaction between melanoma or sarcoma cells with the T-lymphocytes activated by the lysate of selected allogenous melanoma cell lines with high expression of CTA. CONCLUSION Our results demonstrate that one can use lysates from allogeneic melanoma cell lines as a source of CTA for DC load during the production of anticancer vaccines for the STBS treatment. Patterns of CTA expression should be evaluated as biomarkers of response in prospective clinical trials.
Collapse
Affiliation(s)
- Anna Danilova
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
- Department of Oncoimmunology, N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Vsevolod Misyurin
- N.N. Blokhin’ National Medical Cancer Research Center, Kashirskoye sh. 24, Moscow, 115478 Russian Federation
| | - Aleksei Novik
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Dmitry Girdyuk
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Natalia Avdonkina
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Tatiana Nekhaeva
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Natalia Emelyanova
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Nino Pipia
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Andrey Misyurin
- N.N. Blokhin’ National Medical Cancer Research Center, Kashirskoye sh. 24, Moscow, 115478 Russian Federation
| | - Irina Baldueva
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| |
Collapse
|
32
|
Liu Y, Han Y, Dong H, Wei X, Shi D, Li Y. Ca 2+-Mediated Surface Polydopamine Engineering to Program Dendritic Cell Maturation. ACS APPLIED MATERIALS & INTERFACES 2020; 12:4163-4173. [PMID: 31891476 DOI: 10.1021/acsami.9b20997] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Engineering of cell surfaces holds promise in manipulating cellular activities in a physicochemical route as a complement to the biological approach. Mediated by Ca2+, a quick and convenient yet cytocompatible method is used to achieve surface engineering, by which polydopamine nanostructures can be in situ grown onto dendritic cell (DC) surfaces within 10 min. Ca2+, as the physical bridge between the negative cell surface and polydopamine, avoids the direct chemical polymerization of polydopamine onto the cell surface, critically important to maintain the cell viability. As a proof of concept in potential applications, this cell surface engineering shows a good control toward DC maturation. Upon surface polydopamine engineering, bone-marrow-derived DC exhibits a unique bidirectional control of maturation. The polydopamine structure enables effective suppression of DC activation by acting as an efficient scavenger of reactive oxygen species, a key signal during maturation. Conversely, an 808 nm laser irradiation can remotely relieve the suppressed state and effectively activate DC maturation by the photoheat effect of polydopamine (39 °C). The work provides an easily implemented, straightforward approach to achieve cell surface engineering, through which the DC maturation can be controlled.
Collapse
Affiliation(s)
- Yiqiong Liu
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| | - Yi Han
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| | - Haiqing Dong
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| | - Xunbin Wei
- Med-X Research Institute and School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai 200030 , China
| | - Donglu Shi
- The Materials Science & Engineering Program, Department of Mechanical & Materials Engineering, College of Engineering & Applied Science , University of Cincinnati , Cincinnati , Ohio 45221 , United States
| | - Yongyong Li
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| |
Collapse
|
33
|
Guisier F, Barros-Filho MC, Rock LD, Strachan-Whaley M, Marshall EA, Dellaire G, Lam WL. Janus or Hydra: The Many Faces of T Helper Cells in the Human Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1224:35-51. [PMID: 32036603 DOI: 10.1007/978-3-030-35723-8_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CD4+ T helper (TH) cells are key regulators in the tumour immune microenvironment (TIME), mediating the adaptive immunological response towards cancer, mainly through the activation of cytotoxic CD8+ T cells. After antigen recognition and proper co-stimulation, naïve TH cells are activated, undergo clonal expansion, and release cytokines that will define the differentiation of a specific effector TH cell subtype. These different subtypes have different functions, which can mediate both anti- and pro-tumour immunological responses. Here, we present the dual role of TH cells restraining or promoting the tumour, the factors controlling their homing and differentiation in the TIME, their influence on immunotherapy, and their use as prognostic indicators.
Collapse
Affiliation(s)
- Florian Guisier
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada. .,Department of Pneumology, Thoracic Oncology and Intensive Respiratory Care, Rouen University Hospital, Rouen, France.
| | - Mateus Camargo Barros-Filho
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,International Research Center, A.C.Camargo Cancer Center, Sao Paulo, SP, Brazil
| | - Leigha D Rock
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Department of Oral and Biological Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC, Canada.,Department of Cancer Control Research, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Faculty of Dentistry, Dalhousie University, Halifax, NS, Canada
| | | | - Erin A Marshall
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Graham Dellaire
- Department of Pathology, Dalhousie University, Halifax, NS, Canada.,Canadian Environmental Exposures in Cancer (CE2C) Network (CE2C.ca), Halifax, NS, Canada
| | - Wan L Lam
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Canadian Environmental Exposures in Cancer (CE2C) Network (CE2C.ca), Halifax, NS, Canada
| |
Collapse
|
34
|
Fromm PD, Silveira PA, Hsu JL, Papadimitrious MS, Lo TH, Ju X, Kupresanin F, Romano A, Hsu WH, Bryant CE, Kong B, Abadir E, Mekkawy A, M McGuire H, Groth BFDS, Cunningham I, Newman E, Gibson J, Hogarth PM, Hart DNJ, Clark GJ. Distinguishing human peripheral blood CD16 + myeloid cells based on phenotypic characteristics. J Leukoc Biol 2019; 107:323-339. [PMID: 31749181 DOI: 10.1002/jlb.5a1119-362rrr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/28/2022] Open
Abstract
Myeloid lineage cells present in human peripheral blood include dendritic cells (DC) and monocytes. The DC are identified phenotypically as HLA-DR+ cells that lack major cell surface lineage markers for T cells (CD3), B cells (CD19, CD20), NK cells (CD56), red blood cells (CD235a), hematopoietic stem cells (CD34), and Mo that express CD14. Both DC and Mo can be phenotypically divided into subsets. DC are divided into plasmacytoid DC, which are CD11c- , CD304+ , CD85g+ , and myeloid DC that are CD11c+ . The CD11c+ DC are readily classified as CD1c+ DC and CD141+ DC. Monocytes are broadly divided into the CD14+ CD16- (classical) and CD14dim CD16+ subsets (nonclassical). A population of myeloid-derived cells that have DC characteristics, that is, HLA-DR+ and lacking lineage markers including CD14, but express CD16 are generally clustered with CD14dim CD16+ monocytes. We used high-dimensional clustering analyses of fluorescence and mass cytometry data, to delineate CD14+ monocytes, CD14dim CD16+ monocytes (CD16+ Mo), and CD14- CD16+ DC (CD16+ DC). We sought to identify the functional and kinetic relationship of CD16+ DC to CD16+ Mo. We demonstrate that differentiation of CD16+ DC and CD16+ Mo during activation with IFNγ in vitro and as a result of an allo-hematopoietic cell transplant (HCT) in vivo resulted in distinct populations. Recovery of blood CD16+ DC in both auto- and allo-(HCT) patients after myeloablative conditioning showed similar reconstitution and activation kinetics to CD16+ Mo. Finally, we show that expression of the cell surface markers CD300c, CCR5, and CLEC5a can distinguish the cell populations phenotypically paving the way for functional differentiation as new reagents become available.
Collapse
Affiliation(s)
- Phillip D Fromm
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Pablo A Silveira
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Jennifer L Hsu
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia
| | - Michael S Papadimitrious
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Tsun-Ho Lo
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Xinsheng Ju
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Fiona Kupresanin
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia
| | - Adelina Romano
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Department of Pathology, The University of Sydney, Sydney, New South Wales, Australia
| | - Wei-Hsun Hsu
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Christian E Bryant
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Kong
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Edward Abadir
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Ahmed Mekkawy
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia
| | - Helen M McGuire
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Department of Pathology, The University of Sydney, Sydney, New South Wales, Australia
| | - Barbara Fazekas de St Groth
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Department of Pathology, The University of Sydney, Sydney, New South Wales, Australia
| | - Ilona Cunningham
- Department of Haematology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Elizabeth Newman
- Department of Haematology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - John Gibson
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - P Mark Hogarth
- Immune Therapies Group, Burnet Institute, Melbourne, Victoria, Australia
| | - Derek N J Hart
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Institute of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Georgina J Clark
- Dendritic Cell Research, ANZAC Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
35
|
Sprooten J, Ceusters J, Coosemans A, Agostinis P, De Vleeschouwer S, Zitvogel L, Kroemer G, Galluzzi L, Garg AD. Trial watch: dendritic cell vaccination for cancer immunotherapy. Oncoimmunology 2019; 8:e1638212. [PMID: 31646087 PMCID: PMC6791419 DOI: 10.1080/2162402x.2019.1638212] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Dendritic- cells (DCs) have received considerable attention as potential targets for the development of anticancer vaccines. DC-based anticancer vaccination relies on patient-derived DCs pulsed with a source of tumor-associated antigens (TAAs) in the context of standardized maturation-cocktails, followed by their reinfusion. Extensive evidence has confirmed that DC-based vaccines can generate TAA-specific, cytotoxic T cells. Nonetheless, clinical efficacy of DC-based vaccines remains suboptimal, reflecting the widespread immunosuppression within tumors. Thus, clinical interest is being refocused on DC-based vaccines as combinatorial partners for T cell-targeting immunotherapies. Here, we summarize the most recent preclinical/clinical development of anticancer DC vaccination and discuss future perspectives for DC-based vaccines in immuno-oncology.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jolien Ceusters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
- Department of Gynecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
- Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Steven De Vleeschouwer
- Research Group Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
- Université de Paris Descartes, Paris, France
| | - Abhishek D. Garg
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
36
|
Krakow S, Crescimone ML, Bartels C, Wiegering V, Eyrich M, Schlegel PG, Wölfl M. Re-expression of CD14 in Response to a Combined IL-10/TLR Stimulus Defines Monocyte-Derived Cells With an Immunoregulatory Phenotype. Front Immunol 2019; 10:1484. [PMID: 31316520 PMCID: PMC6611188 DOI: 10.3389/fimmu.2019.01484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/13/2019] [Indexed: 01/10/2023] Open
Abstract
Interleukin 10 is a central regulator of the antigen-presenting function of myeloid cells. It exerts immunomodulatory effects in vivo and induces a regulatory phenotype in monocyte-derived cells in vitro. We analyzed phenotype and function of monocytic cells in vitro in relation to the cytokine milieu and the timing of TLR-based activation. In GM-CSF/IL-4 cultured human monocytic cells, we identified two, mutually exclusive cell populations arising from undifferentiated cells: CD83+ fully activated dendritic cells and CD14+ macrophage like cells. Re-expression of CD14 occurs primarily after a sequential trigger with a TLR signal following IL-10 preincubation. This cell population with re-expressed CD14 greatly differs in phenotype and function from the CD83+ cells. Detailed analysis of individual subpopulations reveals that exogenous IL-10 is critical for inducing the shift toward the CD14+ population, but does not affect individual changes in marker expression or cell function in most cases. Thus, plasticity of CD14 expression, defining a subset of immunoregulatory cells, is highly relevant for the composition of cellular products (such as DC vaccines) as it affects the function of the total product.
Collapse
Affiliation(s)
- Sören Krakow
- Department of Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Marie L Crescimone
- Department of Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Charlotte Bartels
- Department of Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Verena Wiegering
- Department of Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Matthias Eyrich
- Department of Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Paul G Schlegel
- Department of Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Matthias Wölfl
- Department of Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital, University of Würzburg, Würzburg, Germany
| |
Collapse
|
37
|
Zhao W, Hou X, Vick OG, Dong Y. RNA delivery biomaterials for the treatment of genetic and rare diseases. Biomaterials 2019; 217:119291. [PMID: 31255978 DOI: 10.1016/j.biomaterials.2019.119291] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/14/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022]
Abstract
Genetic and rare diseases (GARDs) affect more than 350 million patients worldwide and remain a significant challenge in the clinic. Hence, continuous efforts have been made to bridge the significant gap between the supply and demand of effective treatments for GARDs. Recent decades have witnessed the impressive progress in the fight against GARDs, with an improved understanding of the genetic origins of rare diseases and the rapid development in gene therapy providing a new avenue for GARD therapy. RNA-based therapeutics, such as RNA interference (RNAi), messenger RNA (mRNA) and RNA-involved genome editing technologies, demonstrate great potential as a therapy tool for treating genetic associated rare diseases. In the meantime, a variety of RNA delivery vehicles were established for boosting the widespread applications of RNA therapeutics. Among all the RNA delivery platforms which enable the systemic applications of RNAs, non-viral RNA delivery biomaterials display superior properties and a few biomaterials have been successfully exploited for achieving the RNA-based gene therapies on GARDs. In this review article, we focus on recent advances in the development of novel biomaterials for delivery of RNA-based therapeutics and highlight their applications to treat GARDs.
Collapse
Affiliation(s)
- Weiyu Zhao
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, United States
| | - Xucheng Hou
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, United States
| | - Olivia G Vick
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, United States; Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, United States; The Center for Clinical and Translational Science, The Ohio State University, Columbus, OH, 43210, United States; The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, United States; Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, 43210, United States; Department of Radiation Oncology, The Ohio State University, Columbus, OH, 43210, United States.
| |
Collapse
|
38
|
Wang L, Fan J, Ye W, Han J, Zhang Y, Zhao L, Duan J, Yin D, Yi Y. The Expression of ILT4 in Myeloid Dendritic Cells in Patients with Hepatocellular Carcinoma. Immunol Invest 2019; 48:704-718. [PMID: 31044626 DOI: 10.1080/08820139.2019.1571507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Immunoglobulin-like transcript (ILT) 4 is an inhibitory immune receptor of the immunoglobulin superfamily, which could deliver inhibitory signals and induce immunosuppression. The significance of the expression of ILT4 in mDCs subsets in patients with hepatocellular carcinoma (HCC) remains unclear. In this study, the frequency of mDCs subsets in the peripheral blood of 121 patients with HCC and 103 normal controls, and in the tumor and tumor free liver tissues (TFL) of 43 HCC patients was analyzed by flow cytometry. Then, the expressions of ILT4 in mDCs subsets in the microenvironment of liver cancer were also analyzed. Results showed that the percentage of CD1c+ subset was dramatically decreased in peripheral blood mononuclear cells (PBMCs) of HCC patients compared with normal controls, and also significantly decreased in tumor tissue compared with the TFL. The decreased of CD1c+ subset in blood could be a diagnostic factor for HCC with the area under the receiver operating characteristic curve 0.975 (P < 0.01). The percentage of ILT4+CD1c+ subset was dramatically increased in tumor than that of TFL and blood. There were significant correlations between the percentage of ILT4+ in CD1c+ subset in tumor and that of in blood. The percentage of ILT4+CD1c+ subset in tumor tissue was strongly associated with the Edmondson-Steiner stage in HCC (P = 0.03). Furthermore, the capacity of ILT4+CD1c+ subset producing IFN-γ was lower than ILT4- CD1c subset in PBMC of HCC patients following Poly I:C stimulation. Taken together, the increased ILT4+CD1c+ subset in tumor tissue might play an important role in immune suppression for patients with HCC.
Collapse
Affiliation(s)
- Lili Wang
- Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine , Nanjing , PR China
| | - Jing Fan
- Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine , Nanjing , PR China
| | - Wei Ye
- Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine , Nanjing , PR China
| | - Jianbo Han
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine , Nanjing , PR China
| | - Yufeng Zhang
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine , Nanjing , PR China
| | - Liang Zhao
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine , Nanjing , PR China
| | - Jie Duan
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine , Nanjing , PR China
| | - Dandan Yin
- Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine , Nanjing , PR China
| | - Yongxiang Yi
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine , Nanjing , PR China
| |
Collapse
|
39
|
Kather JN, Halama N. Harnessing the innate immune system and local immunological microenvironment to treat colorectal cancer. Br J Cancer 2019; 120:871-882. [PMID: 30936499 PMCID: PMC6734657 DOI: 10.1038/s41416-019-0441-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/20/2019] [Accepted: 03/05/2019] [Indexed: 12/14/2022] Open
Abstract
Significant progress in the development of new immunotherapies has led to successful clinical trials for malignant melanoma and non-small cell lung cancer; however, for the majority of solid tumours of the gastrointestinal tract, little or no progress has been seen. The efficacy of immunotherapies is limited by the complexities of a diverse set of immune cells, and interactions between the tumour cells and all other cells in the local microenvironment of solid tumours. A large fraction of immune cells present in and around solid tumours derive from the innate arm of the immune system and using these cells against tumours offers an alternative immunotherapeutic option, especially as current strategies largely harness the adaptive arm of the immune system. This option is currently being investigated and attempts at using the innate immune system for gastrointestinal cancers are showing initial results. Several important factors, including cytokines, chemotherapeutics and the microbiome, influence the plasticity and functionality of innate (myeloid) cells in the microenvironment, and this complexity of regulation has limited translation into successful trials so far. In this review, current concepts of the immunobiology of the innate arm in the tumour microenvironment are presented in the context of clinical translation.
Collapse
Affiliation(s)
- Jakob Nikolas Kather
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,German Translational Cancer Consortium (DKTK), Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany. .,Institute for Immunology, University Hospital Heidelberg, Heidelberg, Germany. .,Department of Translational Immunotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Helmholtz Institute for Translational Oncology (HI-TRON), Mainz, Germany.
| |
Collapse
|
40
|
da Silva LT, Santillo BT, de Almeida A, Duarte AJDS, Oshiro TM. Using Dendritic Cell-Based Immunotherapy to Treat HIV: How Can This Strategy be Improved? Front Immunol 2018; 9:2993. [PMID: 30619346 PMCID: PMC6305438 DOI: 10.3389/fimmu.2018.02993] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/04/2018] [Indexed: 11/13/2022] Open
Abstract
Harnessing dendritic cells (DC) to treat HIV infection is considered a key strategy to improve anti-HIV treatment and promote the discovery of functional or sterilizing cures. Although this strategy represents a promising approach, the results of currently published trials suggest that opportunities to optimize its performance still exist. In addition to the genetic and clinical characteristics of patients, the efficacy of DC-based immunotherapy depends on the quality of the vaccine product, which is composed of precursor-derived DC and an antigen for pulsing. Here, we focus on some factors that can interfere with vaccine production and should thus be considered to improve DC-based immunotherapy for HIV infection.
Collapse
Affiliation(s)
- Laís Teodoro da Silva
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Bruna Tereso Santillo
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alexandre de Almeida
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alberto Jose da Silva Duarte
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Telma Miyuki Oshiro
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
41
|
Dionisi M, De Archangelis C, Battisti F, Rahimi Koshkaki H, Belleudi F, Zizzari IG, Ruscito I, Albano C, Di Filippo A, Torrisi MR, Benedetti Panici P, Napoletano C, Nuti M, Rughetti A. Tumor-Derived Microvesicles Enhance Cross-Processing Ability of Clinical Grade Dendritic Cells. Front Immunol 2018; 9:2481. [PMID: 30455687 PMCID: PMC6230586 DOI: 10.3389/fimmu.2018.02481] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 10/08/2018] [Indexed: 12/29/2022] Open
Abstract
Tumor cells release extracellular microvesicles (MVs) in the microenvironment to deliver biological signals to neighboring cells as well as to cells in distant tissues. Tumor-derived MVs appear to play contradictory role promoting both immunosuppression and tumor growth and both evoking tumor specific immune response. Recent evidences indicate that tumor-derived MVs can positively impact Dendritic Cells (DCs) immunogenicity by reprogramming DC antigen processing machinery and intracellular signaling pathways, thus promoting anti-tumor response. DCs are considered pivot cells of the immune system due to their exclusive ability to coordinate the innate and acquired immune responses, cross-present exogenous antigens, and prime naïve T cells. DCs are required for the induction and maintenance of long-lasting anti-tumor immunity and their exploitation has been extensively investigated for the design of anti-tumor vaccines. However, the clinical grade culture conditions that are required to generate DCs for therapeutic use can strongly affect their functions. Here, we investigated the immunomodulatory impact of MVs carrying the MUC1 tumor glycoantigen (MVsMUC1) as immunogen formulation on clinical grade DCs grown in X-VIVO 15 (X-DCs). Results indicated that X-DCs displayed reduced performance of the antigen processing machinery in term of diminished phagocytosis and acidification of the phagosomal compartment suggesting an altered immunogenicity of clinical grade DCs. Pulsing DCs with MVsMUC1 restored phagosomal alkalinization, triggering ROS increase. This was not observed when a soluble MUC1 protein was employed (rMUC1). Concurrently, MVsMUC1 internalization by X-DCs allowed MUC1 cross-processing. Most importantly, MVsMUC1 pulsed DCs activated IFNγ response mediated by MUC1 specific CD8+ T cells. These results strongly support the employment of tumor-derived MVs as immunogen platforms for the implementation of DC-based vaccines.
Collapse
Affiliation(s)
- Marco Dionisi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | | | - Federico Battisti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | | | - Francesca Belleudi
- Department of Clinical and Molecular Medicine, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, "Sapienza" University of Rome, Rome, Italy
| | | | - Ilary Ruscito
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.,European Competence Center for Ovarian Cancer, Department of Gynecology, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Christian Albano
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | | | - Maria Rosaria Torrisi
- Department of Clinical and Molecular Medicine, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, "Sapienza" University of Rome, Rome, Italy.,U.O.C. Genetica medica e Diagnostica cellulare avanzata, S. Andrea University Hospital, Rome, Italy
| | | | - Chiara Napoletano
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Marianna Nuti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Aurelia Rughetti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
42
|
Raaijmakers TK, Ansems M. Microenvironmental derived factors modulating dendritic cell function and vaccine efficacy: the effect of prostanoid receptor and nuclear receptor ligands. Cancer Immunol Immunother 2018; 67:1789-1796. [PMID: 29998375 PMCID: PMC6208817 DOI: 10.1007/s00262-018-2205-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/09/2018] [Indexed: 01/20/2023]
Abstract
Dendritic cells (DCs) are widely used in DC-based immunotherapies because of their capacity to steer immune responses. So far treatment success is limited and more functional knowledge on how DCs initiate and stably drive specific responses is needed. Many intrinsic and extrinsic factors contribute to how DCs skew the immune response towards immunity or tolerance. The origin and type of DC, its maturation status, but also factors they encounter in the in vitro or in vivo microenvironment they reside in during differentiation and maturation affect this balance. Treatment success of DC vaccines will, therefore, also depend on the presence of these factors during the process of vaccination. Identification and further knowledge of natural and pharmacological compounds that modulate DC differentiation and function towards a specific response may help to improve current DC-based immunotherapies. This review focuses on factors that could improve the efficacy of DC vaccines in (pre-)clinical studies to enhance DC-based immunotherapy, with a particular emphasis on compounds acting on prostanoid or nuclear receptor families.
Collapse
Affiliation(s)
- Tonke K Raaijmakers
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, The Netherlands
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
43
|
Wefers C, Schreibelt G, Massuger LFAG, de Vries IJM, Torensma R. Immune Curbing of Cancer Stem Cells by CTLs Directed to NANOG. Front Immunol 2018; 9:1412. [PMID: 29971070 PMCID: PMC6018198 DOI: 10.3389/fimmu.2018.01412] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/06/2018] [Indexed: 12/15/2022] Open
Abstract
Cancer stem cells (CSCs) have been identified as the source of tumor growth and disease recurrence. Eradication of CSCs is thus essential to achieve durable responses, but CSCs are resistant to current anti-tumor therapies. Novel therapeutic approaches that specifically target CSCs will, therefore, be crucial to improve patient outcome. Immunotherapies, which boost the body's own immune system to eliminate cancerous cells, could be an alternative approach to target CSCs. Vaccines of dendritic cells (DCs) loaded with tumor antigens can evoke highly specific anti-tumor T cell responses. Importantly, DC vaccination also promotes immunological memory formation, paving the way for long-term cancer control. Here, we propose a DC vaccination that specifically targets CSCs. DCs loaded with NANOG peptides, a protein required for maintaining stem cell properties, could evoke a potent anti-tumor immune response against CSCs. We hypothesize that the resulting immunological memory will also control newly formed CSCs, thereby preventing disease recurrence.
Collapse
Affiliation(s)
- Christina Wefers
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
- Department of Obstetrics and Gynecology, Radboudumc, Nijmegen, Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
| | | | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
| | - Ruurd Torensma
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
| |
Collapse
|
44
|
Pan P, Huang YW, Oshima K, Yearsley M, Zhang J, Yu J, Arnold M, Wang LS. An immunological perspective for preventing cancer with berries. JOURNAL OF BERRY RESEARCH 2018; 8:163-175. [PMID: 30159104 PMCID: PMC6110394 DOI: 10.3233/jbr-180305] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Berries and their phytochemicals have well documented chemopreventive roles, but understanding their ability to regulate cancer immunology is only beginning to be explored. The literature, including human studies, suggests that berry components can modulate our immune system to delay cancer development. Moreover, their wide spectrum of phytochemicals suggests that they might influence the functions of multiple immune cells and different aspects of cancer immunity. Cancer immune-therapies are showing promise for some types of cancer because they boost T cells' ability to recognize tumor cells - an essential prelude to destruction. Recognition occurs after dendritic cells present antigen, such as tumor antigen, to T cells, generating an adaptive response. Therefore, the potential of berries to aid cancer immune-therapies by, for example, regulating dendritic cells, warrants further investigation in animal and human studies. More information is also needed about berries' effects on the entire spectrum of immunity so that a comprehensive view can inform efforts to use berries to enhance immune responses during cancer prevention and treatment. This review summarizes the effects of berries as anti-tumor agents from the immunological perspective in tumor-bearing animals and humans.
Collapse
Affiliation(s)
- Pan Pan
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yi-Wen Huang
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kiyoko Oshima
- Department of Pathology, John Hopkins University, Baltimore, MD, USA
| | - Martha Yearsley
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Jianying Zhang
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Jianhua Yu
- Department of Internal Medicine, Division of Hematology, College of Medicine, Comprehensive Cancer Center and The James Cancer Hospital, The Ohio State University, Columbus, OH, USA
| | - Mark Arnold
- Department of Surgery, The Ohio State University, OH, USA
| | - Li-Shu Wang
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
- Corresponding author: Li-Shu Wang, Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, RM C4930, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA. Tel.: +1 414 955 2827; Fax: +1 414 955 6059; .
| |
Collapse
|