1
|
Gordy JT, Hui Y, Schill C, Wang T, Chen F, Fessler K, Meza J, Li Y, Taylor AD, Bates RE, Karakousis PC, Pekosz A, Sachithanandham J, Li M, Karanika S, Markham RB. A SARS-CoV-2 RBD vaccine fused to the chemokine MIP-3α elicits sustained murine antibody responses over 12 months and enhanced lung T-cell responses. Front Immunol 2024; 15:1292059. [PMID: 38370404 PMCID: PMC10870766 DOI: 10.3389/fimmu.2024.1292059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 01/15/2024] [Indexed: 02/20/2024] Open
Abstract
Background Previous studies have demonstrated enhanced efficacy of vaccine formulations that incorporate the chemokine macrophage inflammatory protein 3α (MIP-3α) to direct vaccine antigens to immature dendritic cells. To address the reduction in vaccine efficacy associated with a mutation in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mutants, we have examined the ability of receptor-binding domain vaccines incorporating MIP-3α to sustain higher concentrations of antibody when administered intramuscularly (IM) and to more effectively elicit lung T-cell responses when administered intranasally (IN). Methods BALB/c mice aged 6-8 weeks were immunized intramuscularly or intranasally with DNA vaccine constructs consisting of the SARS-CoV-2 receptor-binding domain alone or fused to the chemokine MIP-3α. In a small-scale (n = 3/group) experiment, mice immunized IM with electroporation were followed up for serum antibody concentrations over a period of 1 year and for bronchoalveolar antibody levels at the termination of the study. Following IN immunization with unencapsulated plasmid DNA (n = 6/group), mice were evaluated at 11 weeks for serum antibody concentrations, quantities of T cells in the lungs, and IFN-γ- and TNF-α-expressing antigen-specific T cells in the lungs and spleen. Results At 12 months postprimary vaccination, recipients of the IM vaccine incorporating MIP-3α had significantly, approximately threefold, higher serum antibody concentrations than recipients of the vaccine not incorporating MIP-3α. The area-under-the-curve analyses of the 12-month observation interval demonstrated significantly greater antibody concentrations over time in recipients of the MIP-3α vaccine formulation. At 12 months postprimary immunization, only recipients of the fusion vaccine had concentrations of serum-neutralizing activity deemed to be effective. After intranasal immunization, only recipients of the MIP-3α vaccine formulations developed T-cell responses in the lungs significantly above those of PBS controls. Low levels of serum antibody responses were obtained following IN immunization. Conclusion Although requiring separate IM and IN immunizations for optimal immunization, incorporating MIP-3α in a SARS-CoV-2 vaccine construct demonstrated the potential of a stable and easily produced vaccine formulation to provide the extended antibody and T-cell responses that may be required for protection in the setting of emerging SARS-CoV-2 variants. Without electroporation, simple, uncoated plasmid DNA incorporating MIP-3α administered intranasally elicited lung T-cell responses.
Collapse
Affiliation(s)
- James Tristan Gordy
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Yinan Hui
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Courtney Schill
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Tianyin Wang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Fengyixin Chen
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Kaitlyn Fessler
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Jacob Meza
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Yangchen Li
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Alannah D Taylor
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Rowan E Bates
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Petros C Karakousis
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Jaiprasath Sachithanandham
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Maggie Li
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Styliani Karanika
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Richard B Markham
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
2
|
Kudling TV, Clubb JH, Pakola S, Quixabeira DC, Lähdeniemi IA, Heiniö C, Arias V, Havunen R, Cervera-Carrascon V, Santos JM, Sutinen E, Räsänen J, Borenius K, Mäyränpää MI, Aaltonen E, Sorsa S, Hemminki O, Kanerva A, Verschuren EW, Ilonen I, Hemminki A. Effective intravenous delivery of adenovirus armed with TNFα and IL-2 improves anti-PD-1 checkpoint blockade in non-small cell lung cancer. Oncoimmunology 2023; 12:2241710. [PMID: 37546696 PMCID: PMC10399490 DOI: 10.1080/2162402x.2023.2241710] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023] Open
Abstract
Lung cancer remains among the most difficult-to-treat malignancies and is the leading cause of cancer-related deaths worldwide. The introduction of targeted therapies and checkpoint inhibitors has improved treatment outcomes; however, most patients with advanced-stage non-small cell lung cancer (NSCLC) eventually fail these therapies. Therefore, there is a major unmet clinical need for checkpoint refractory/resistant NSCLC. Here, we tested the combination of aPD-1 and adenovirus armed with TNFα and IL-2 (Ad5-CMV-mTNFα/mIL-2) in an immunocompetent murine NSCLC model. Moreover, although local delivery has been standard for virotherapy, treatment was administered intravenously to facilitate clinical translation and putative routine use. We showed that treatment of tumor-bearing animals with aPD-1 in combination with intravenously injected armed adenovirus significantly decreased cancer growth, even in the presence of neutralizing antibodies. We observed an increased frequency of cytotoxic tumor-infiltrating lymphocytes, including tumor-specific cells. Combination treatment led to a decreased percentage of immunosuppressive tumor-associated macrophages and an improvement in dendritic cell maturation. Moreover, we observed expansion of the tumor-specific memory T cell compartment in secondary lymphoid organs in the group that received aPD-1 with the virus. However, although the non-replicative Ad5-CMV-mTNFα/mIL-2 virus allows high transgene expression in the murine model, it does not fully reflect the clinical outcome in humans. Thus, we complemented our findings using NSCLC ex vivo models fully permissive for the TNFα and IL-2- armed oncolytic adenovirus TILT-123. Overall, our data demonstrate the ability of systemically administered adenovirus armed with TNFα and IL-2 to potentiate the anti-tumor efficacy of aPD-1 and warrant further investigation in clinical trials.
Collapse
Affiliation(s)
- Tatiana V. Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - James H.A. Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dafne C.A. Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Iris A.K. Lähdeniemi
- Translational Lung Cancer Research Group, Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Joao M. Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Eva Sutinen
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pulmonary Medicine, Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Jari Räsänen
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristian Borenius
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko I. Mäyränpää
- Pathology, University of Helsinki and Helsinki University Hospital (HUSLAB), Helsinki, Finland
| | - Eero Aaltonen
- Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Otto Hemminki
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Gynecology and Obstetrics, Helsinki University Hospital, Helsinki, Finland
| | - Emmy W. Verschuren
- Translational Lung Cancer Research Group, Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Ilkka Ilonen
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
| |
Collapse
|
3
|
Baran K, Kordiak J, Jabłoński S, Brzeziańska-Lasota E. Panel of miR-150 and linc00673, regulators of CCR6/CCL20 may serve as non-invasive diagnostic marker of non-small cell lung cancer. Sci Rep 2023; 13:9642. [PMID: 37316552 DOI: 10.1038/s41598-023-36485-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/05/2023] [Indexed: 06/16/2023] Open
Abstract
The C-C motif ligand 20 (CCL20) is a chemokine that specifically binds to the chemokine receptor 6 (CCR6) and the CCL20/CCR6 axis has been implicated in the non-small lung cancer (NSCLC) development and progression. Its expression is regulated by mutual interactions of non-coding RNAs (ncRNAs). This goals of presented study was to evaluate the expression level of CCR6/CCL20 mRNA in NSCLC tissue comparative to selected ncRNAs: miR-150, linc00673. The expression level of the studied ncRNAs was also assessed in serum extracellular vesicles (EVs). Thirty patients (n = 30) were enrolled as the study cohort. Total RNA was isolated from tumor tissue, adjacent macroscopically unchanged tissue and serum EVs. The expression level of studied genes and ncRNAs were estimated based on the qPCR method. Higher expression level of CCL20 mRNA but lower expression level of CCR6 mRNA were observed in tumor in comparison to control tissue. Relative to the smoking status, higher CCL20 (p < 0.05) and CCR6 mRNA (p > 0.05) expression levels were observed in current smokers than in never smokers. In serum EVs the expression level of miR-150 has a negative correlation with AJCC tumor staging, whereas the expression level of linc00673 positively correlated (p > 0.05). The lower expression level of miR-150 and higher expression level of linc00673 in serum EVs were observed in NSCLC patients with lymph nodes metastases (p > 0.05). Regarding the histopathological type, significantly lower expression level of miR-150 and higher expression level of linc00673 were observed in the serum EVs of patients with AC compared to patient with SCC. Our findings revealed that smoking significantly changed the expression level of CCL20 mRNA in NSCLC tissue. Changes in expression levels of miR-150 and linc00673 in the serum EVs of NSCLC patients in relation to presence of lymph node metastases and the stage of cancer development may serve as a non-invasive molecular biomarkers of tumor progression. Furthermore, expression levels of miR-150 and linc00673 may serve as non-intrusive diagnostic biomarkers differentiating adenocarcinoma from squamous cell carcinoma.
Collapse
Affiliation(s)
- Kamila Baran
- Department of Biomedicine and Genetics, Chair of Biology and Medical Microbiology, Medical University of Lodz, Lodz, Poland.
| | - Jacek Kordiak
- Department of Thoracic, General and Oncological Surgery, Medical University of Lodz, Lodz, Poland
| | - Sławomir Jabłoński
- Department of Thoracic, General and Oncological Surgery, Medical University of Lodz, Lodz, Poland
| | - Ewa Brzeziańska-Lasota
- Department of Biomedicine and Genetics, Chair of Biology and Medical Microbiology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
4
|
Savage TM, Vincent RL, Rae SS, Huang LH, Ahn A, Pu K, Li F, de los Santos-Alexis K, Coker C, Danino T, Arpaia N. Chemokines expressed by engineered bacteria recruit and orchestrate antitumor immunity. SCIENCE ADVANCES 2023; 9:eadc9436. [PMID: 36888717 PMCID: PMC9995032 DOI: 10.1126/sciadv.adc9436] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 02/07/2023] [Indexed: 05/28/2023]
Abstract
Tumors use multiple mechanisms to actively exclude immune cells involved in antitumor immunity. Strategies to overcome these exclusion signals remain limited due to an inability to target therapeutics specifically to the tumor. Synthetic biology enables engineering of cells and microbes for tumor-localized delivery of therapeutic candidates previously unavailable using conventional systemic administration techniques. Here, we engineer bacteria to intratumorally release chemokines to attract adaptive immune cells into the tumor environment. Bacteria expressing an activating mutant of the human chemokine CXCL16 (hCXCL16K42A) offer therapeutic benefit in multiple mouse tumor models, an effect mediated via recruitment of CD8+ T cells. Furthermore, we target the presentation of tumor-derived antigens by dendritic cells, using a second engineered bacterial strain expressing CCL20. This led to type 1 conventional dendritic cell recruitment and synergized with hCXCL16K42A-induced T cell recruitment to provide additional therapeutic benefit. In summary, we engineer bacteria to recruit and activate innate and adaptive antitumor immune responses, offering a new cancer immunotherapy strategy.
Collapse
Affiliation(s)
- Thomas M. Savage
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Rosa L. Vincent
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Sarah S. Rae
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Lei Haley Huang
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Alexander Ahn
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Kelly Pu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Fangda Li
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | | | - Courtney Coker
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Tal Danino
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Data Science Institute, Columbia University, New York, NY, USA
| | - Nicholas Arpaia
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| |
Collapse
|
5
|
Stuckel AJ, Khare T, Bissonnette M, Khare S. Aberrant regulation of CXCR4 in cancer via deviant microRNA-targeted interactions. Epigenetics 2022; 17:2318-2331. [PMID: 36047714 PMCID: PMC9665135 DOI: 10.1080/15592294.2022.2118947] [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: 03/25/2022] [Revised: 08/02/2022] [Accepted: 08/18/2022] [Indexed: 11/03/2022] Open
Abstract
CXCR4 is involved in many facets of cancer, including being a major player in establishing metastasis. This is in part due to the deregulation of CXCR4, which can be attributed to many genetic and epigenetic mechanisms, including aberrant microRNA-CXCR4 interaction. MicroRNAs (miRNAs) are a type of small non-coding RNA that primarily targets the 3' UTR of mRNA transcripts, which in turn suppresses mRNA and subsequent protein expression. In this review, we reported and characterized the many aberrant miRNA-CXCR4 interactions that occur throughout human cancers. In particular, we reported known target sequences located on the 3' UTR of CXCR4 transcripts that tumour suppressor miRNAs bind and therefore regulate expression by. From these aberrant interactions, we also documented affected downstream genes/pathways and whether a particular tumour suppressor miRNA was reported as a prognostic marker in its respected cancer type. In addition, a limited number of cancer-causing miRNAs coined 'oncomirs' were reported and described in relation to CXCR4 regulation. Moreover, the mechanisms underlying both tumour suppressor and oncomir deregulations concerning CXCR4 expression were also explored. Furthermore, the miR-146a-CXCR4 axis was delineated in oncoviral infected endothelial cells in the context of virus-causing cancers. Lastly, miRNA-driven therapies and CXCR4 antagonist drugs were discussed as potential future treatment options in reported cancers pertaining to deregulated miRNA-CXCR4 interactions.
Collapse
Affiliation(s)
- Alexei J. Stuckel
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri65212, USA
| | - Tripti Khare
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri65212, USA
| | - Marc Bissonnette
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago, Chicago, Il60637, USA
| | - Sharad Khare
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri65212, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri65201, USA
| |
Collapse
|
6
|
Fu G, Wu Y, Zhao G, Chen X, Xu Z, Sun J, Tian J, Cheng Z, Shi Y, Jin B. Activation of cGAS-STING Signal to Inhibit the Proliferation of Bladder Cancer: The Immune Effect of Cisplatin. Cells 2022; 11:cells11193011. [PMID: 36230972 PMCID: PMC9564335 DOI: 10.3390/cells11193011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/09/2022] [Accepted: 09/22/2022] [Indexed: 11/29/2022] Open
Abstract
Cisplatin is commonly used in neoadjuvant, adjuvant, and systemic therapy for advanced bladder cancer, but its immune-related mechanism is still unclear. Exploration of the immune effects of cisplatin in bladder cancer would complement the comprehensive mechanism of cisplatin and provide the basis for combination therapy of cisplatin and immunotherapy in bladder cancer. We confirmed the immune effects of cisplatin on T24 and TCCSUP bladder cancer cell lines in vitro and explored the important function of these immune effects in the bladder cancer microenvironment in a mice tumor model. We found cisplatin induced immune response in bladder cancer by RNA sequencing and validated that cGAS-STING signal was deeply involved in this response. Cisplatin induced cGAS-STING signal inhibited the proliferation of bladder cancer and increased the infiltration percentages of CD8+ T cells and dendritic cells in a transplantation mice tumor model. Accumulation of dsDNA and the release of chromatin bound cGAS are important to activate downstream STING. Our findings indicated a cisplatin-related immune effect in bladder cancer, and cisplatin combined with immunotherapy might have a synergistic effect for bladder cancer therapy.
Collapse
|
7
|
Huang FY, Wang JY, Dai SZ, Lin YY, Sun Y, Zhang L, Lu Z, Cao R, Tan GH. A recombinant oncolytic Newcastle virus expressing MIP-3α promotes systemic antitumor immunity. J Immunother Cancer 2021; 8:jitc-2019-000330. [PMID: 32759233 PMCID: PMC7410001 DOI: 10.1136/jitc-2019-000330] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The oncolytic Newcastle disease virus (NDV) is inherently able to trigger the lysis of tumor cells and induce the immunogenic cell death (ICD) of tumor cells and is also an excellent gene-engineering vector. The macrophage inflammatory protein-3α (MIP-3α) is a specific chemokine for dendritic cells (DCs). Thus, we constructed a recombinant NDV expressing MIP-3α (NDV-MIP3α) as an in vivo DC vaccine for amplifying antitumor immunities. METHODS The recombinant NDV-MIP3α was constructed by the insertion of MIP-3α cDNA between the P and M genes. Western blotting assay and ELISA were used to detect MIP-3α, HMGB1, IgG, and ATP in the supernatant and sera. The chemotaxis of DCs was examined by Transwell chambers. The phenotypes of the immune cells (eg, DCs) were analyzed by flow cytometry. The antitumor efficiency of NDV-MIP3α was observed in B16 and CT26 tumor-bearing mice. Immunofluorescence and immunohistochemistry were applied to observe the ecto-calreticulin (CRT) and intratumoral attraction of DCs. Adoptive transfer of splenocytes and antibodies and depletion of T-cell subsets were used to evaluate the relationship between antitumor immunities and the role of the T-cell subtype. RESULTS The findings show that NDV-MIP3α has almost the same capabilities of tumor lysis and induction of ICD as the wild-type NDV (NDV-WT). MIP-3α secreted by NDV-MIP3α could successfully attract DCs in vitro and in vivo. Both B16 and CT26 cells infected with NDV-MIP3α could strongly promote DC maturation and activation. Compared with NDV-WT, intratumoral injection of NDV-MIP3α and the adoptive transfer of T lymphocytes from mice injected with NDV-MIP3α resulted in a significant suppression of B16 and CT26 tumor growth. The NDV-MIP3α-induced production of tumor-specific cellular and humoral immune responses was dependent on CD8+ T cells and partially on CD4+ T cells. A significant reversion of tumor microenvironments was found in the mice injected with NDV-MIP3α. CONCLUSIONS Compared with NDV-WT, the recombinant NDV-MIP3α as an in vivo DC vaccine demonstrates enhanced antitumor activities through the induction of stronger system immunities and modulation of the tumor microenvironment. This strategy may be a potential approach for the generation of an in vivo DC vaccine.
Collapse
Affiliation(s)
- Feng-Ying Huang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Jin-Yan Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Shu-Zhen Dai
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Ying-Ying Lin
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Yan Sun
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Liming Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Zhuoxuan Lu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Rong Cao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Guang-Hong Tan
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
8
|
De Zutter A, Van Damme J, Struyf S. The Role of Post-Translational Modifications of Chemokines by CD26 in Cancer. Cancers (Basel) 2021; 13:cancers13174247. [PMID: 34503058 PMCID: PMC8428238 DOI: 10.3390/cancers13174247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Chemokines are a large family of small chemotactic cytokines that fulfill a central function in cancer. Both tumor-promoting and -impeding roles have been ascribed to chemokines, which they exert in a direct or indirect manner. An important post-translational modification that regulates chemokine activity is the NH2-terminal truncation by peptidases. CD26 is a dipeptidyl peptidase (DPPIV), which typically clips a NH2-terminal dipeptide from the chemokine. With a certain degree of selectivity in terms of chemokine substrate, CD26 only recognizes chemokines with a penultimate proline or alanine. Chemokines can be protected against CD26 recognition by specific amino acid residues within the chemokine structure, by oligomerization or by binding to cellular glycosaminoglycans (GAGs). Upon truncation, the binding affinity for receptors and GAGs is altered, which influences chemokine function. The consequences of CD26-mediated clipping vary, as unchanged, enhanced, and reduced activities are reported. In tumors, CD26 most likely has the most profound effect on CXCL12 and the interferon (IFN)-inducible CXCR3 ligands, which are converted into receptor antagonists upon truncation. Depending on the tumor type, expression of CD26 is upregulated or downregulated and often results in the preferential generation of the chemokine isoform most favorable for tumor progression. Considering the tight relationship between chemokine sequence and chemokine binding specificity, molecules with the appropriate characteristics can be chemically engineered to provide innovative therapeutic strategies in a cancer setting.
Collapse
|
9
|
Zhao H, Chen C, Chen X, Yang C, Zhang D, Li Y, Zhao H, He J. The Collective Effect of MIP-3α and FL Promotes Dendritic Cell Function Within the Immune Microenvironment of Murine Liver Cancer. Front Oncol 2021; 11:646527. [PMID: 33842360 PMCID: PMC8032989 DOI: 10.3389/fonc.2021.646527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/09/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma is a highly malignant and lethal tumor. In addition to surgery, immunotherapy is currently a more effective treatment for hepatocellular carcinoma. The tumor immune microenvironment (TIME) largely determines the efficacy of cancer immunotherapy. Based on the universal targeting of TIME modulators in clinical treatment, TIME modulators are promising targets for tumor immunotherapy. We investigated the effect of a double gene expression vector (recombinant galactose-terminal glycol-poly-L-lysine coupled MIP-3α-FL) on dendritic cells (DCs) regulation within the TIME of mice with liver cancer. H22 cells were transfected with a recombinant MIP-3α-FL plasmid to induce DCs differentiation and chemotaxis. The effects of transfection were investigated by flow cytometry following the modified Boyden’s method. Cytokine-induced killer (CIK) cells co-culture revealed changes in the antigen presentation ability of DCs. Further, tumor-bearing mice were injected with the recombinant double gene vector via the tail vein. We compared the survival time, tumor volume, weight of the mice, as well as the number and phenotype of tumor-infiltrating DCs (TIDCs) between groups. The supernatant of transfected H22 cells promoted the phenotypic maturation of DCs, enhancing their chemotaxis. Further, treated DCs promoted the cytokine secretion and killing ability of CIK cells. The survival time of mice injected with the double gene vector was significantly prolonged, while their tumor weight and volume were relatively reduced. Flow cytometry revealed that the number of TIDCs (as well as CD80 and CD86 expression) in the MouseMIP-3α-FL group, were significantly higher than in the control group. The combination of MIP-3α and FL can significantly promote DCs aggregation, maturation, and enhance their antigen presentation ability. The coupling of the double gene vector with glycosylated polylysine can improve the precise targeting of the liver and inhibit tumor growth in vivo, providing a novel approach for immune therapy in liver cancer.
Collapse
Affiliation(s)
- Haichao Zhao
- Graduate School, Shanxi Medical University, Taiyuan, China.,Department of Hepatobiliary Surgery, Shanxi Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Changzhou Chen
- Graduate School, Shanxi Medical University, Taiyuan, China
| | - Xidong Chen
- Graduate School, Shanxi Medical University, Taiyuan, China
| | - Chuanli Yang
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Donglin Zhang
- Graduate School, Shanxi Medical University, Taiyuan, China
| | - Yanjun Li
- Graduate School, Shanxi Medical University, Taiyuan, China.,Department of Hepatobiliary Surgery, Shanxi Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Haoliang Zhao
- Graduate School, Shanxi Medical University, Taiyuan, China.,Department of Hepatobiliary Surgery, Shanxi Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Jiefeng He
- Graduate School, Shanxi Medical University, Taiyuan, China.,Department of Hepatobiliary Surgery, Shanxi Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
10
|
Abstract
Recent advances in immunotherapy have revolutionized the treatment of certain cancers. Some patients show a durable response to these immunotherapies, while others show little benefit or develop resistance. Identification of biomarkers to predict responsiveness will be helpful for informing treatment strategies; and would furthermore lead to the identification of molecular pathways dysregulated in nonresponding patients that could be targeted for therapeutic development. Pathways of epigenetic modification, such as histone posttranslational modifications (PTMs), have been shown to be dysregulated in certain cancer and immune cells. Histones are abundant cellular proteins readily assayed with high-throughput technologies, making them attractive targets as biomarkers. We explore promising advancements for using histone PTMs as immunotherapy responsiveness biomarkers in both cancer and immune cells, and provide a methodological workflow for assaying histone PTMs in relevant samples.
Collapse
Affiliation(s)
- Erin M Taylor
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Brian Koss
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lauren E Davis
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
11
|
Da Silva CG, Camps MGM, Li TMWY, Chan AB, Ossendorp F, Cruz LJ. Co-delivery of immunomodulators in biodegradable nanoparticles improves therapeutic efficacy of cancer vaccines. Biomaterials 2019; 220:119417. [PMID: 31419588 DOI: 10.1016/j.biomaterials.2019.119417] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/11/2019] [Accepted: 08/06/2019] [Indexed: 01/08/2023]
Abstract
To improve the efficacy of cancer vaccines we aimed to modulate the suppressive tumor microenvironment. In this study, the potential of intratumoral immune modulation with poly (I:C), Resiquimod (R848) and CCL20 (MIP3α) was explored. Biodegradable polymeric nanoparticles were used as delivery vehicles for slow and sustained release of these drugs in the tumor area and were combined with specific immunotherapy based on therapeutic peptide vaccination in two aggressive murine carcinoma and lymphoma tumor models. Whereas nanoparticle delivery of poly (I:C) or R848 improved therapeutic efficacy, the combination with MIP3α remarkably potentiated the cancer vaccine antitumor effects. The long-term survival increased to 75-100% and the progression free survival nearly doubled on mice with established large carcinoma tumors. The potent adjuvant effects were associated with lymphoid and myeloid population alterations in the tumor and tumor-draining lymph node. In addition to a significant influx of macrophages into the tumor, the phenotype of the suppressor tumor-associated macrophages shifted towards an acute inflammatory phenotype in the tumor-draining lymph node. Overall, these data show that therapeutic cancer vaccines can be potentiated by the combined nanoparticle mediated co-delivery of poly (I:C), R848 and MIP3α, which indicates that a more favorable milieu for cancer fighting immune cells is created for T cells induced by therapeutic cancer vaccines.
Collapse
Affiliation(s)
- C G Da Silva
- Department of Radiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - M G M Camps
- Department of Immunohematology and Blood Transfusion, LUMC, Leiden, the Netherlands
| | - T M W Y Li
- Department of Radiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - A B Chan
- Department of Radiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands; Percuros B.V., Enschede, the Netherlands
| | - F Ossendorp
- Department of Immunohematology and Blood Transfusion, LUMC, Leiden, the Netherlands
| | - L J Cruz
- Department of Radiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands.
| |
Collapse
|
12
|
Yam AO, Chtanova T. The Ins and Outs of Chemokine-Mediated Immune Cell Trafficking in Skin Cancer. Front Immunol 2019; 10:386. [PMID: 30899263 PMCID: PMC6416210 DOI: 10.3389/fimmu.2019.00386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/14/2019] [Indexed: 12/14/2022] Open
Abstract
Recent studies of the patterns of chemokine-mediated immune cell recruitment into solid tumors have enhanced our understanding of the role played by various immune cell subsets both in amplifying and inhibiting tumor cell growth and spread. Here we discuss how the chemokine/chemokine receptor networks bring together immune cells within the microenvironment of skin tumors, particularly melanomas, including their effect on disease progression, prognosis and therapeutic options.
Collapse
Affiliation(s)
- Andrew O. Yam
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Tatyana Chtanova
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
13
|
Mantovani A, Schioppa T, Biswas SK, Marchesi F, Allavena P, Sica A. Tumor-Associated Macrophages and Dendritic Cells as Prototypic Type II Polarized Myeloid Populations. TUMORI JOURNAL 2018; 89:459-68. [PMID: 14870765 DOI: 10.1177/030089160308900501] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Environmental signals polarize mononuclear phagocytes which can express different functional programmes. Fully polarized type I and type II (or alternatively activated) macrophages are the extremes of a continuum of functional states. Tumor-derived and T cell-derived cytokines stimulate tumor associated macrophages (TAM) to acquire a polarized type II phenotype. These functionally polarized cells, and similarly oriented or immature dendritic cells present in tumors, play a key role in subversion of adaptive immunity and in inflammatory circuits which promote tumor growth and progression.
Collapse
|
14
|
Huang FY, Lei J, Sun Y, Yan F, Chen B, Zhang L, Lu Z, Cao R, Lin YY, Wang CC, Tan GH. Induction of enhanced immunogenic cell death through ultrasound-controlled release of doxorubicin by liposome-microbubble complexes. Oncoimmunology 2018; 7:e1446720. [PMID: 29900064 DOI: 10.1080/2162402x.2018.1446720] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 02/23/2018] [Accepted: 02/23/2018] [Indexed: 02/07/2023] Open
Abstract
Immunogenic cell death (ICD) is a specific kind of cell death that stimulates the immune system to combat cancer cells. Ultrasound (US)-controlled targeted release of drugs by liposome-microbubble complexes is a promising approach due to its non-invasive nature and visibility through ultrasound imaging. However, it is not known whether this approach can enhance ICD induced by drugs, such as doxorubicin. Herein, we prepared a doxorubicin-liposome-microbubble complex (MbDox), and the resultant MbDox was then characterized and tested for US-controlled release of Dox (MbDox+US treatment) to enhance the induction of ICD in LL/2 and CT26 cancer cells and in syngeneic murine models. We found that MbDox+US treatment caused more cellular uptake and nuclear accumulation of Dox in tumor cells, and more accumulation of Dox in tumor tissues. Enhanced induction of ICD occurred both in vitro and in vivo. MbDox+US treatment induced more apoptosis, stronger membrane exposure and the release of ER stress proteins and DAMPs in tumor cells, and increased DC maturation in vitro. In addition, MbDox+US treatment also resulted in stronger therapeutic effects in immunocompetent mice than in immunodeficient mice. Moreover, MbDox+US enhancement of ICD was also evidenced by a higher proportion of activated CD8+ T-lymphocytes but lower Treg in tumor tissues. Taken together, our results demonstrate that US-controlled release of ICD inducers into nuclei using liposome-microbubble complexes may be an effective approach to enhance the induction of ICD for tumor treatment.
Collapse
Affiliation(s)
- Feng-Ying Huang
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Jing Lei
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China.,Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical College, Haikou , China
| | - Yan Sun
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Fei Yan
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Bin Chen
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Liming Zhang
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Zhuoxuan Lu
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Rong Cao
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Ying-Ying Lin
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Cai-Chun Wang
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China.,Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical College, Haikou , China
| | - Guang-Hong Tan
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| |
Collapse
|
15
|
Rippaus N, Taggart D, Williams J, Andreou T, Wurdak H, Wronski K, Lorger M. Metastatic site-specific polarization of macrophages in intracranial breast cancer metastases. Oncotarget 2018; 7:41473-41487. [PMID: 27203741 PMCID: PMC5173073 DOI: 10.18632/oncotarget.9445] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/10/2016] [Indexed: 11/25/2022] Open
Abstract
In contrast to primary tumors, the understanding of macrophages within metastases is very limited. In order to compare macrophage phenotypes between different metastatic sites, we established a pre-clinical mouse model of intracranial breast cancer metastasis in which cancer lesions develop simultaneously within the brain parenchyma and the dura. This mimics a situation that is commonly occurring in the clinic. Flow cytometry analysis revealed significant differences in the activation state of metastasis-associated macrophages (MAMs) at the two locations. Concurrently, gene expression analysis identified significant differences in molecular profiles of cancer cells that have metastasized to the brain parenchyma as compared to the dura. This included differences in inflammation-related pathways, NF-kB1 activity and cytokine profiles. The most significantly upregulated cytokine in brain parenchyma- versus dura-derived cancer cells was Lymphotoxin β and a gain-of-function approach demonstrated a direct involvement of this factor in the M2 polarization of parenchymal MAMs. This established a link between metastatic site-specific properties of cancer cells and the MAM activation state.
Collapse
Affiliation(s)
- Nora Rippaus
- Institute of Cancer and Pathology, University of Leeds, St. James's University Hospital, LS9 7TF Leeds, UK
| | - David Taggart
- Institute of Cancer and Pathology, University of Leeds, St. James's University Hospital, LS9 7TF Leeds, UK
| | - Jennifer Williams
- Institute of Cancer and Pathology, University of Leeds, St. James's University Hospital, LS9 7TF Leeds, UK
| | - Tereza Andreou
- Institute of Cancer and Pathology, University of Leeds, St. James's University Hospital, LS9 7TF Leeds, UK
| | - Heiko Wurdak
- Institute of Cancer and Pathology, University of Leeds, St. James's University Hospital, LS9 7TF Leeds, UK
| | | | - Mihaela Lorger
- Institute of Cancer and Pathology, University of Leeds, St. James's University Hospital, LS9 7TF Leeds, UK
| |
Collapse
|
16
|
Sathianathen NJ, Krishna S, Anderson JK, Weight CJ, Gupta S, Konety BR, Griffith TS. The current status of immunobased therapies for metastatic renal-cell carcinoma. Immunotargets Ther 2017; 6:83-93. [PMID: 29255699 PMCID: PMC5723125 DOI: 10.2147/itt.s134850] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The management of metastatic renal-cell carcinoma (mRCC) represents an important clinical challenge. Since being approved in the early 1990s, aspecific immunotherapy has been a mainstay of treatment for mRCC and the only therapy that has demonstrated long-term cures for mRCC. However, in recent times there have been landmark advances made in the field of specific immunotherapy for a number of malignancies, including kidney cancer. This review outlines the range of immunobased agents currently available for the treatment of mRCC.
Collapse
Affiliation(s)
| | | | | | | | - Shilpa Gupta
- Masonic Cancer Center
- Division of Hematology, Oncology, and Transplantation
| | | | - Thomas S Griffith
- Department of Urology
- Masonic Cancer Center
- Center for Immunology
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
17
|
Schaller TH, Batich KA, Suryadevara CM, Desai R, Sampson JH. Chemokines as adjuvants for immunotherapy: implications for immune activation with CCL3. Expert Rev Clin Immunol 2017; 13:1049-1060. [PMID: 28965431 DOI: 10.1080/1744666x.2017.1384313] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Immunotherapy embodies any approach that manipulates the immune system for therapeutic benefit. In this regard, various clinical trials have employed direct vaccination with patient-specific dendritic cells or adoptive T cell therapy to target highly aggressive tumors. Both modalities have demonstrated great specificity, an advantage that is unmatched by other treatment strategies. However, their full potential has yet to be realized. Areas covered: In this review, we provide an overview of chemokines in pathogen and anti-tumor immune responses and discuss further improving immunotherapies by arming particular chemokine axes. Expert commentary: The chemokine macrophage inflammatory protein-1 alpha (MIP-1α, CCL3) has emerged as a potent activator of both innate and adaptive responses. Specifically, CCL3 plays a critical role in recruiting distinct immune phenotypes to intratumoral sites, is a pivotal player in regulating lymph node homing of dendritic cell subsets, and induces antigen-specific T cell responses. The recent breadth of literature outlines the various interactions of CCL3 with these cellular subsets, which have now served as a basis for immunotherapeutic translation.
Collapse
Affiliation(s)
- Teilo H Schaller
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Kristen A Batich
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Carter M Suryadevara
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Rupen Desai
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA
| | - John H Sampson
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b Department of Pathology , Duke University Medical Center , Durham , NC , USA.,c Department of Radiation Oncology , Duke University Medical Center , Durham , NC , USA.,d Department of Immunology , Duke University Medical Center , Durham , NC , USA
| |
Collapse
|
18
|
Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol 2017; 17:559-572. [PMID: 28555670 DOI: 10.1038/nri.2017.49] [Citation(s) in RCA: 1490] [Impact Index Per Article: 186.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The tumour microenvironment is the primary location in which tumour cells and the host immune system interact. Different immune cell subsets are recruited into the tumour microenvironment via interactions between chemokines and chemokine receptors, and these populations have distinct effects on tumour progression and therapeutic outcomes. In this Review, we focus on the main chemokines that are found in the human tumour microenvironment; we elaborate on their patterns of expression, their regulation and their roles in immune cell recruitment and in cancer and stromal cell biology, and we consider how they affect cancer immunity and tumorigenesis. We also discuss the potential of targeting chemokine networks, in combination with other immunotherapies, for the treatment of cancer.
Collapse
Affiliation(s)
- Nisha Nagarsheth
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, Michigan 48109, USA.,Graduate Programs in Immunology and Tumour Biology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Max S Wicha
- Graduate Programs in Immunology and Tumour Biology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Medicine, University of Michigan School of Medicine, 1150 E. Medical Center Drive, Ann Arbor, Michigan 48109, USA.,The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, Michigan 48109, USA.,Graduate Programs in Immunology and Tumour Biology, University of Michigan, Ann Arbor, Michigan 48109, USA.,The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
19
|
Wang B, Shi L, Sun X, Wang L, Wang X, Chen C. Production of CCL20 from lung cancer cells induces the cell migration and proliferation through PI3K pathway. J Cell Mol Med 2016; 20:920-9. [PMID: 26968871 PMCID: PMC4831357 DOI: 10.1111/jcmm.12781] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/07/2015] [Indexed: 11/28/2022] Open
Abstract
Tumour inflammatory microenvironment is considered to play a role in the sensitivity of tumour cells to therapies and prognosis of patients with lung cancer. The expression of CCL20, one of the critical chemoattractants responsible for inflammation cells recruitment, has been shown overexpressed in variety of tumours. This study aimed at investigating potential mechanisms of CCL20 function and production in human non-small cell lung cancer (NSCLC). Expression of CCL20 gene and protein in lung tissues of patients with NSCLC and NSCLC cells (A549) were determined. The interleukin (IL)-1β-induced signal pathways in A549 and the effect of CCL20-induced A549 cell migration and proliferation were determined using migration assays and cell-alive monitoring system. Mechanisms of signal pathways involved in the migration of CCL20 were also studied. We initially found that NSCLC tumour tissues markedly overexpressed CCL20 in comparison with normal lung samples. In addition, IL-1β could directly promote CCL20 production in lung cancer cells, which was inhibited by extracellular signal-regulated kinase (ERK)1/2 inhibitor, p38 mitogen-activated protein kinase (p38 MARP) inhibitor or PI3K inhibitors. CCL20 promoted lung cancer cells migration and proliferation in an autocrine manner via activation of ERK1/2-MAPK and PI3K pathways. Our data indicated that IL-1β could stimulate CCL20 production from lung cancer cells through the activation of MAPKs and PI3K signal pathways, and the auto-secretion of CCL20 could promote lung cancer cell migration and proliferation through the activation of ERK and PI3K signal pathways. Our results may provide a novel evidence that CCL20 could be a new therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Beibei Wang
- Department of Lung Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lin Shi
- Zhongshan Hospital Biomedical Research Center, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiaoru Sun
- Department of Lung Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lingyan Wang
- Zhongshan Hospital Biomedical Research Center, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiangdong Wang
- Department of Lung Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
- Zhongshan Hospital Biomedical Research Center, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Fudan University Shanghai Medical College, Shanghai, China
| | - Chengshui Chen
- Department of Lung Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
20
|
Huang FY, Huang FR, Chen B, Liu Q, Wang H, Zhou SL, Zhao HG, Huang YH, Lin YY, Tan GH. Microencapsulation of tumor lysates and live cell engineering with MIP-3α as an effective vaccine. Biomaterials 2015; 53:554-65. [PMID: 25890751 DOI: 10.1016/j.biomaterials.2015.02.123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/25/2015] [Accepted: 02/27/2015] [Indexed: 10/23/2022]
Abstract
The combination of several potential strategies so as to develop new tumor vaccines is an attractive field of translational medicine. Pulsing tumor lysates with dendritic cells (DCs), in-vivo attraction of DCs by macrophage inflammatory protein 3α (MIP-3α), and reversion of the tumor suppressive microenvironment have been tested as strategies to develop tumor vaccines. In this study, we generated an alginate microsphere (named PaLtTcAdMIP3α) that encapsulated tumor lysates, live tumor cells engineering with a recombinant MIP-3α adenovirus and BCG. We used PaLtTcAdMIP3α as a model vaccine to test its antitumor activities. Our results showed that PaLtTcAdMIP3α expressed and excreted MIP-3α, which effectively attracted DCs ex vivo and in vivo. Injection of PaLtTcAdMIP3α into tumor-bearing mice effectively induced both therapeutic and prophylactic antitumor immunities in CT26, Meth A, B16-F10 and H22 models, but without any ensuing increase in adverse effects. Both tumor-specific cellular and humoral immune responses, especially the CD8(+) T cell-dependent cytotoxic T immunity, were found in the mice injected with PaLtTcAdMIP3α. The anti-tumor activity was abrogated completely by depletion of CD8(+) and partially by CD4(+) T lymphocytes. In addition, the number of IFN-γ-producing CD8(+) T cells in spleen and tumor tissues was significantly increased; but the number of CD4(+)CD25(+)FOXP3(+) regulatory T cells (Treg) in tumor tissues was decreased. These data strongly suggest that a combination of multi-current-using strategies such as the novel approach of using our PaLtTcAdMIP3α microspheres could be an effective tumor model vaccine.
Collapse
Affiliation(s)
- Feng-ying Huang
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou 571199, China
| | - Feng-ru Huang
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou 571199, China
| | - Bin Chen
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou 571199, China
| | - Quan Liu
- Oncology Institute, Fourth Affiliated Hospital of Soochow University, Wuxi 214062, China
| | - Hua Wang
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou 571199, China
| | - Song-lin Zhou
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou 571199, China
| | - Huan-ge Zhao
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou 571199, China
| | - Yong-hao Huang
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou 571199, China
| | - Ying-ying Lin
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou 571199, China
| | - Guang-hong Tan
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou 571199, China.
| |
Collapse
|
21
|
Sharma RK, Chheda ZS, Jala VR, Haribabu B. Regulation of cytotoxic T-Lymphocyte trafficking to tumors by chemoattractants: implications for immunotherapy. Expert Rev Vaccines 2014; 14:537-49. [PMID: 25482400 DOI: 10.1586/14760584.2015.982101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cancer immunotherapy has recently emerged as an important treatment modality. FDA approval of provenge, ipilimumab and pembrolizumab has started to deliver on the long awaited promise of cancer immunotherapy. Many new modalities of immunotherapies targeting cytotoxic T lymphocytes (CTLs) responses, such as adoptive cell therapies and vaccines, are in advanced clinical trials. In all these immunotherapies, migration of CTLs to the tumor site is a critical step for achieving therapeutic efficacy. However, inefficient infiltration of activated CTLs into established tumors is increasingly being recognized as one of the major hurdles limiting efficacy. Mechanisms that control migration of CTLs to tumors are poorly defined. In this review, the authors discuss the chemoattractants and their receptors that have been implicated in endogenous- or immunotherapy-induced CTL recruitment to tumors and the potential for targeting these pathways for therapeutic efficacy.
Collapse
Affiliation(s)
- Rajesh K Sharma
- James Graham Brown Cancer Center, University of Louisville Health Sciences, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
22
|
Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, Huang J, Zhou Y, Yao X, Bian X, Wang JM. The role of chemoattractant receptors in shaping the tumor microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:751392. [PMID: 25110692 PMCID: PMC4119707 DOI: 10.1155/2014/751392] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Chemoattractant receptors are a family of seven transmembrane G protein coupled receptors (GPCRs) initially found to mediate the chemotaxis and activation of immune cells. During the past decades, the functions of these GPCRs have been discovered to not only regulate leukocyte trafficking and promote immune responses, but also play important roles in homeostasis, development, angiogenesis, and tumor progression. Accumulating evidence indicates that chemoattractant GPCRs and their ligands promote the progression of malignant tumors based on their capacity to orchestrate the infiltration of the tumor microenvironment by immune cells, endothelial cells, fibroblasts, and mesenchymal cells. This facilitates the interaction of tumor cells with host cells, tumor cells with tumor cells, and host cells with host cells to provide a basis for the expansion of established tumors and development of distant metastasis. In addition, many malignant tumors of the nonhematopoietic origin express multiple chemoattractant GPCRs that increase the invasiveness and metastasis of tumor cells. Therefore, GPCRs and their ligands constitute targets for the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Jiamin Zhou
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Endoscopic Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Xiang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Keqiang Chen
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Jian Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ye Zhou
- Department of Gastric Cancer and Soft Tissue Surgery, Fudan University Cancer Center, Shanghai 200032, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
23
|
Modulation of hepatitis C virus core DNA vaccine immune responses by co-immunization with CC-chemokine ligand 20 (CCL20) gene as immunoadjuvant. Mol Biol Rep 2014; 41:5943-52. [PMID: 24972567 DOI: 10.1007/s11033-014-3470-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 06/14/2014] [Indexed: 12/26/2022]
Abstract
Plasmid DNA vaccination is a promising vaccine platform for prevention and treatment of infectious disease. Enhancement of the DNA vaccine potency by co-inoculation of immunoadjuvant has been shown to be an effective strategy. Modulation of dendritic cells and T-cells locomotion and trafficking to prime an immune response is mediated by distinct chemokines. The recent study was designed to elucidate the adjuvant activity of plasmid expressing CC-chemokine ligand 20 (pCCL20) in co-inoculation with hepatitis C virus (HCV) core DNA vaccine immunization. pCCL20 was constructed and evaluated for its functional expression. Sub-cutaneous inoculation of pCCL20 with HCV core DNA vaccine was performed via electroporation in BALB/c mice on day 0 and 14 and a HCV core protein booster was applied on day 28. On week after final immunization, both humoral and cell-mediated immune responses were assessed by indirect ELISA for core specific antibodies, lymphocyte proliferation, cytokine ELISA/ELISpot and cytotoxic Grenzyme B (GrzB) release assays. Mice were co-immunized with pCCL20 developed higher levels of core specific IFN-γ/IL-4 ratio and IL-2 release, IFN-γ producing cells, lymphocyte proliferation and cytotoxic Grenzyme B release in both draining lymph nodes and spleen cells of immunized mice. The core-specific serum total IgG and IgG2a/IgG1 ratio were significantly higher when the pCCL20 was co-inoculated. These results suggest the potential of CCL20 chemokine as vaccine adjuvant to enhance Th1 mediated cellular and humoral immune responses in HCV core DNA immunization.
Collapse
|
24
|
Adenovirus-mediated CCL20/IL-15 gene transfer enhances antitumor immunity in mice. Immunobiology 2014; 219:475-81. [DOI: 10.1016/j.imbio.2014.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 12/29/2013] [Accepted: 02/20/2014] [Indexed: 12/11/2022]
|
25
|
Evaluation of antibody–chemokine fusion proteins for tumor-targeting applications. Exp Biol Med (Maywood) 2014; 239:842-852. [DOI: 10.1177/1535370214536667] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
There is an increasing biotechnological interest in the ‘arming’ of therapeutic antibodies with bioactive payloads. While many antibody–cytokine fusion proteins have been extensively investigated in preclinical and clinical studies, there are only few reports related to antibody–chemokine fusion proteins (‘immunochemokines’). Here, we describe the cloning, expression, and characterization of 10 immunochemokines based on the monoclonal antibody F8, specific to the alternatively spliced extra domain A (EDA) of fibronectin, a marker of angiogenesis. Among the 10 murine chemokines tested in our study, only CCL19, CCL20, CCL21, and CXCL10 could be expressed and isolated at acceptable purity levels as F8-based fusion proteins. The immunochemokines retained the binding characteristics of the parental antibody, but could not be characterized by gel-filtration analysis, an analytical limitation which had previously been observed in our laboratory for the unconjugated chemokines. When radioiodinated preparations of CCL19-F8, CCL20-F8, CCL21-F8, and CXCL10-F8 were tested in quantitative biodistribution studies in tumor-bearing mice, the four fusion proteins failed to preferentially accumulate at the tumor site, while the unconjugated parental antibody displayed a tumor:blood ratio >20:1, 24 h after intravenous (i.v.) administration. The tumor-targeting ability of CCL19-F8 could be rescued only in part by preadministration of unlabeled CCL19-F8, indicating that a chemokine trapping mechanism may hinder pharmacodelivery strategies. While this article highlights expression, analytical, and biodistribution challenges associated with the antibody-based in vivo delivery of chemokines at sites of disease, it provides the first comprehensive report in this field and may facilitate future studies with immunochemokines.
Collapse
|
26
|
Zhao DX, Li ZJ, Zhang Y, Zhang XN, Zhao KC, Li YG, Zhang MM, Yu XW, Liu MY, Li Y. Enhanced antitumor immunity is elicited by adenovirus-mediated gene transfer of CCL21 and IL-15 in murine colon carcinomas. Cell Immunol 2014; 289:155-61. [PMID: 24838092 DOI: 10.1016/j.cellimm.2014.03.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/21/2014] [Accepted: 03/22/2014] [Indexed: 12/19/2022]
Abstract
The chemokine CCL21 is a potent chemoattractant for T cells and dendritic cells. IL-15 elicits powerful antitumor immune responses through the stimulation of natural killer cells. We constructed a CCL21/IL-15-expressing adenovirus (Ad-CCL21-IL-15) and evaluated its antitumor effects in vitro and in vivo. We found that the intratumoral injection of Ad-CCL21-IL-15 into murine colon carcinomas significantly inhibited tumor growth. Splenocytes from mice treated with Ad-CCL21-IL-15 developed tumor-specific cytotoxic T cells and were protected from subsequent challenges with tumor cells. This study indicates that providing cancer therapy by combining CCL21 and IL-15 can induce antitumor immune responses and is an effective strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Dong-xu Zhao
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, China-Japan Union Hospital, Jilin University, Changchun 130062, PR China
| | - Zhi-jie Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, PR China
| | - Yang Zhang
- First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China
| | - Xiao-na Zhang
- First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China
| | - Kun-chi Zhao
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, China-Japan Union Hospital, Jilin University, Changchun 130062, PR China
| | - Ya-gang Li
- Fourth Hospital of Jilin University, Changchun 130062, PR China
| | - Meng-meng Zhang
- Fourth Hospital of Jilin University, Changchun 130062, PR China
| | - Xiao-wei Yu
- First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China
| | - Ming-yuan Liu
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, China-Japan Union Hospital, Jilin University, Changchun 130062, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
| | - Yang Li
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, China-Japan Union Hospital, Jilin University, Changchun 130062, PR China.
| |
Collapse
|
27
|
Abstract
Dendritic cells (DCs) are the most powerful professional antigen-presenting cells and are unique in their capability to initiate, maintain and regulate the intensity of primary immune responses, including specific antitumor responses. Development of practical procedures to prepare sufficient numbers of functional human DCs in culture from the peripheral blood precursors, paved the way for clinical trials to evaluate various DC-based strategies in patients with malignant diseases. However, no definite conclusions regarding the clinical and even immunological efficacy of DC vaccination can be stated, despite the fact that 12 years have passed since the first clinical trial utilizing DCs in cancer patients. Many unanswered questions hamper the development of DC-based vaccines, including the source of DC preparation and protocols for DC generation, activation and loading with tumor antigens, source of tumor antigens, route of vaccine administration and methods of immunomonitoring. Fortunately, in spite of the many obstacles, DC vaccines continue to hold promise for cancer therapy.
Collapse
Affiliation(s)
- Hua Zhong
- Shanghai Jiao Tong University, Shanghai Chest Hospital, 241 Huaihai Road (w), Shanghai 200030, China.
| | | | | |
Collapse
|
28
|
Li D, Wang W, Shi HS, Fu YJ, Chen X, Chen XC, Liu YT, Kan B, Wang YS. Gene therapy with beta-defensin 2 induces antitumor immunity and enhances local antitumor effects. Hum Gene Ther 2013; 25:63-72. [PMID: 24134464 DOI: 10.1089/hum.2013.161] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Beta-defensins, small antimicrobial peptides, are involved in host immune responses to tumors. In this study, we used beta-defensin 2 (BD2) to explore the possible role of beta-defensins in cancer gene therapy. A recombinant plasmid expressing a secretable form of BD2 was constructed. The biological activities of BD2 in immature dendritic cells (iDCs) were tested in vitro and in vivo. The antitumor effects were investigated in three established tumor models. The secreted BD2 was detected and exhibited chemotactic activity in iDCs both in vitro and in vivo. Recruitment and activation of iDCs in tumor niches resulted in significant tumor growth inhibition. Adoptive transfer of splenocytes and depletion of immune cell subsets revealed that CD8(+) T lymphocyte responses mediated the increased tumor inhibition. Furthermore, we also found that chemotactic and maturation-inducing activities in iDCs in tumor milieu contributed to enhanced local antitumor effects. Our study indicates that gene therapy with BD2 can mediate specific antitumor immunity and augment local antitumor effects. Our study also suggested that beta-defensins may merit further exploration for cancer immunotherapy as promising immunogenes.
Collapse
Affiliation(s)
- Dan Li
- Department of Thoracic Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu 610042, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Bobanga ID, Petrosiute A, Huang AY. Chemokines as Cancer Vaccine Adjuvants. Vaccines (Basel) 2013; 1:444-62. [PMID: 24967094 PMCID: PMC4067044 DOI: 10.3390/vaccines1040444] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 08/31/2013] [Accepted: 09/26/2013] [Indexed: 02/07/2023] Open
Abstract
We are witnessing a new era of immune-mediated cancer therapies and vaccine development. As the field of cancer vaccines advances into clinical trials, overcoming low immunogenicity is a limiting step in achieving full success of this therapeutic approach. Recent discoveries in the many biological roles of chemokines in tumor immunology allow their exploitation in enhancing recruitment of antigen presenting cells (APCs) and effector cells to appropriate anatomical sites. This knowledge, combined with advances in gene therapy and virology, allows researchers to employ chemokines as potential vaccine adjuvants. This review will focus on recent murine and human studies that use chemokines as therapeutic anti-cancer vaccine adjuvants.
Collapse
Affiliation(s)
- Iuliana D. Bobanga
- Departments of General Surgery, School of Medicine, University Hospital Case Medical Center/Case Western Reserve University, Cleveland, OH 44106, USA
| | - Agne Petrosiute
- Departments of Pediatrics, School of Medicine, University Hospital Case Medical Center/Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alex Y. Huang
- Departments of Pediatrics, School of Medicine, University Hospital Case Medical Center/Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
30
|
Arab S, Mojarrad M, Motamedi M, Mirzaei R, Modarressi MH, Hadjati J. Tumour regression induced by co-administration of MIP-3α and CpG in an experimental model of colon carcinoma. Scand J Immunol 2013; 78:28-34. [PMID: 23672351 DOI: 10.1111/sji.12058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/25/2013] [Indexed: 01/19/2023]
Abstract
CCL20/macrophage inflammatory protein-3α (MIP-3α) represents one of the potent chemoattractive proteins for dendritic cells (DCs). Herein, we investigated whether in vivo genetic modification of tumour cells aimed at intratumoural production of MIP-3α might lead to accumulation of DCs in tumour tissue. Mice injected with CT26, received recombinant adenovirus (Ad) vectors (AdMIP-3α) expressing MIP-3α protein. This was complemented by injections of CpG. Interestingly, MIP-3α gene therapy combined with CpG injections resulted in specific cytotoxicity. This was associated with significant suppression of tumour growth rate. These findings demonstrate the potential of strategies that utilize in vivo overexpression of chemokines.
Collapse
Affiliation(s)
- S Arab
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
31
|
Yang X, Hou J, Han Z, Wang Y, Hao C, Wei L, Shi Y. One cell, multiple roles: contribution of mesenchymal stem cells to tumor development in tumor microenvironment. Cell Biosci 2013; 3:5. [PMID: 23336752 PMCID: PMC3693909 DOI: 10.1186/2045-3701-3-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 11/20/2012] [Indexed: 12/13/2022] Open
Abstract
The discovery of tissue reparative and immunosuppressive abilities of mesenchymal stem cells (MSCs) has drawn more attention to tumor microenvironment and its role in providing the soil for the tumor cell growth. MSCs are recruited to tumor which is referred as the never healing wound and altered by the inflammation environment, thereby helping to construct the tumor microenvironment. The environment orchestrated by MSCs and other factors can be associated with angiogenesis, immunosuppression, inhibition of apoptosis, epithelial-mesenchymal transition (EMT), survival of cancer stem cells, which all contribute to tumor growth and progression. In this review, we will discuss how MSCs are recruited to the tumor microenvironment and what effects they have on tumor progression.
Collapse
Affiliation(s)
- Xue Yang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, the Second Military Medicial University, 225 Changhai Road, Shanghai 200438, China.
| | | | | | | | | | | | | |
Collapse
|
32
|
Franciszkiewicz K, Boissonnas A, Boutet M, Combadière C, Mami-Chouaib F. Role of chemokines and chemokine receptors in shaping the effector phase of the antitumor immune response. Cancer Res 2012; 72:6325-32. [PMID: 23222302 DOI: 10.1158/0008-5472.can-12-2027] [Citation(s) in RCA: 180] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Immune system-mediated eradication of neoplastic cells requires induction of a strong long-lasting antitumor T-cell response. However, generation of tumor-specific effector T cells does not necessarily result in tumor clearance. CTL must first be able to migrate to the tumor site, infiltrate the tumor tissue, and interact with the target to finally trigger effector functions indispensable for tumor destruction. Chemokines are involved in circulation, homing, retention, and activation of immunocompetent cells. Although some of them are known to contribute to tumor growth and metastasis, others are responsible for changes in the tumor microenvironment that lead to extensive infiltration of lymphocytes, resulting in tumor eradication. Given their chemoattractive and activating properties, a role for chemokines in the development of the effector phase of the antitumor immune response has been suggested. Here, we emphasize the role of the chemokine-chemokine receptor network at multiple levels of the T-cell-mediated antitumor immune response. The identification of chemokine-dependent molecular mechanisms implicated in tumor-specific CTL trafficking, retention, and regulation of their in situ effector functions may offer new perspectives for development of innovative immunotherapeutic approaches to cancer treatment.
Collapse
Affiliation(s)
- Katarzyna Franciszkiewicz
- Institut National de la Santé et de la Recherche Medicale U753, Team 1: Tumor Antigens and T-cell Reactivity, Integrated Research Cancer Institute in Villejuif, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | | | | | | | | |
Collapse
|
33
|
Wang L, Qin H, Li L, Zhang Y, Tu Y, Feng F, Ji P, Zhang J, Li G, Zhao Z, Gao G. Overexpression of CCL20 and its receptor CCR6 predicts poor clinical prognosis in human gliomas. Med Oncol 2012; 29:3491-7. [PMID: 22926920 DOI: 10.1007/s12032-012-0314-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Accepted: 07/16/2012] [Indexed: 12/16/2022]
Abstract
Recent studies have demonstrated that the chemokine CCL20 and its receptor CCR6 may be involved in tumorigenesis, tumor progression and metastatic spread of various human malignancies. The aim of this study was to investigate the clinicopathological significance and prognostic value of CCL20 and CCR6 expression in human malignant glioma. CCL20 and CCR6 expression in human gliomas and nonneoplastic brain tissues was measured by immunohistochemistry. The association of CCL20 and CCR6 expression with clinicopathological factors or prognosis in glioma patients was statistically analyzed. The expression levels of CCL20 and CCR6 proteins were both up-regulated in glioma tissues. There was a significantly positive correlation between the expression of the two markers (r = 0.88; P < 0.001). In addition, the overexpressions of CCL20 and CCR6 were both detected in high-grade glioma tissues compared with those in low-grade tissues and increased with ascending tumor World Health Organization (WHO) grades (P = 0.006 and 0.008, respectively). The increased expressions of CCL20 and CCR6 proteins were also significantly correlated with low Karnofsky performance score (both P = 0.01). Moreover, univariate analysis found that CCL20 expression (P = 0.002), CCR6 expression (P = 0.002) and CCL20/CCR6 co-expression (P < 0.001) were all significantly associated with poor prognosis. In particular, glioma patients with CCL20/CCR6 co-expression have the shortest overall survival. Multivariate analysis further identified the expression levels of CCL20 and CCR6 to be independent prognostic factors. Our data suggest for the first time that CCL20 and CCR6 might play an important role in the regulation of aggressiveness in human gliomas. The up-regulation of CCL20 and CCR6 might be closely associated with poor clinical outcome of patients with gliomas.
Collapse
Affiliation(s)
- Liang Wang
- Department of Neurosurgery, Tangdu Hospital, No. 569, Xinsi Road, Baqiao District, Xi'an City, 710038, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Tissue-Specific Homing of Immune Cells in Malignant Skin Tumors. Pathol Oncol Res 2012; 18:749-59. [DOI: 10.1007/s12253-012-9529-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 03/27/2012] [Indexed: 01/09/2023]
|
35
|
Ding X, Wang K, Wang H, Zhang G, Liu Y, Yang Q, Chen W, Hu S. High expression of CCL20 is associated with poor prognosis in patients with hepatocellular carcinoma after curative resection. J Gastrointest Surg 2012; 16:828-36. [PMID: 22072303 DOI: 10.1007/s11605-011-1775-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Accepted: 10/26/2011] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Chemokine ligand 20 (CCL20) plays an important role in the carcinogenesis, invasion, and progression of malignancies. This study aimed to investigate the prognostic significance of CCL20 in patients with hepatocellular carcinoma (HCC) after curative resection. METHODS Expression of CCL20 was evaluated by immunohistochemistry in tumor tissue from 125 patients who underwent curative resection of HCC. The relationship between CCL20 expression and clinicopathologic features was analyzed. Univariate and multivariate analyses were performed to evaluate its predictive value for tumor recurrence and survival of HCC patients. RESULTS CCL20 expression was verified in all tumor specimens. CCL20 expression was associated with tumor size (P = 0.002), tumor number (P = 0.031), vascular invasion (P = 0.003), tumor differentiation (P = 0.024), and tumor recurrence (P < 0.001). Patients with high CCL20 expression had poorer recurrence-free survival and overall survival (both P < 0.001) than those with low CCL20 expression. Multivariate analysis showed CCL20 expression was an independent predictor of tumor recurrence (Hazard ratio 3.934, P = 0.002), recurrence-free survival (Hazard ratio 2.573, P = 0.001), and overall survival (Hazard ratio 2.930, P = 0.001). CONCLUSION CCL20 expression was associated with tumor recurrence and survival of HCC patients. It may be used to predict prognosis of patients with HCC and may be a new target of postoperative adjuvant therapy.
Collapse
Affiliation(s)
- Xiangjiu Ding
- Department of General Surgery, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Luzina IG, Atamas SP. CCR6 is not necessary for functional effects of human CCL18 in a mouse model. FIBROGENESIS & TISSUE REPAIR 2012; 5:2. [PMID: 22257697 PMCID: PMC3274466 DOI: 10.1186/1755-1536-5-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 01/18/2012] [Indexed: 11/10/2022]
Abstract
CCL18, a chemokine with no known receptor, has been implicated in several fibrotic pulmonary diseases associated with T-lymphocyte infiltration. It has been hypothesized that CCL18 may act through CCR6. Gene delivery of human CCL18 to the lungs of wild-type mice induced pulmonary infiltration of T-lymphocytes, less than 5% of which expressed CCR6. In the lungs of CCR6-deficient mice, CCL18-driven infiltration of T-lymphocytes was attenuated but not fully abrogated. It was concluded that CCR6 is not necessary for CCL18-induced changes in mice in vivo and that CCR6 is not the main functional receptor for CCL18 in this model.
Collapse
Affiliation(s)
- Irina G Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | |
Collapse
|
37
|
Codelivery of the chemokine CCL3 by an adenovirus-based vaccine improves protection from retrovirus infection. J Virol 2011; 86:1706-16. [PMID: 22090142 DOI: 10.1128/jvi.06244-11] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Processing and presentation of vaccine antigens by professional antigen-presenting cells (APCs) is of great importance for the efficient induction of protective immunity. We analyzed whether the efficacy of an adenovirus-based retroviral vaccine can be enhanced by coadministration of adenovirus-encoded chemokines that attract and stimulate APCs. In the Friend retrovirus (FV) mouse model we coexpressed CCL3, CCL20, CCL21, or CXCL14 from adenoviral vectors, together with FV Gag and Env antigens, and then analyzed immune responses and protection from pathogenic FV infection. Although most tested chemokines did not improve protection against FV challenge, mice that received adenoviral vectors encoding CCL3 together with FV antigens showed significantly better control over viral loads and FV-induced disease than mice immunized with the viral antigens only. Improved protection correlated with enhanced virus-specific CD4+ T cell responses and higher neutralizing antibody titers. To apply these results to an HIV vaccine, mice were immunized with adenoviral vectors encoding the HIV antigens Env and Gag-Pol and coadministered vectors encoding CCL3. Again, this combination vaccine induced higher virus-specific antibody titers and CD4+ T cell responses than did the HIV antigens alone. These results indicate that coexpression of the chemokine CCL3 by adenovirus-based vectors may be a promising tool to improve antiretroviral vaccination strategies.
Collapse
|
38
|
Raman D, Sobolik-Delmaire T, Richmond A. Chemokines in health and disease. Exp Cell Res 2011; 317:575-89. [PMID: 21223965 DOI: 10.1016/j.yexcr.2011.01.005] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Accepted: 01/03/2011] [Indexed: 12/20/2022]
Abstract
Chemokines and their receptors play a key role in development and homeostasis as well as in the pathogenesis of tumors and autoimmune diseases. Chemokines are involved in the implantation of the early conceptus, the migration of subsets of cells during embryonic development, and the overall growth of the embryo. Chemokines also have an important role in the development and maintenance of innate and adaptive immunity. In addition, they play a significant role in wound healing and angiogenesis. When the physiological role of chemokines is subverted or chronically amplified, disease often follows. Chemokines are involved in the pathobiology of chronic inflammation, tumorigenesis and metastasis, as well as autoimmune diseases. This article reviews the role of chemokines and their receptors in normal and disease processes and the potential for using chemokine antagonists for appropriate targeted therapy.
Collapse
Affiliation(s)
- Dayanidhi Raman
- Department of Cancer Biology, Vanderbilt University, School of Medicine, USA
| | | | | |
Collapse
|
39
|
Chen D, Yang K, Zhang G, Mei J, Xiang L. Screen and analysis of key disease genes for precancerous lesions of oral buccal mucosa induced by DMBA in golden hamsters. Oncol Lett 2010; 2:265-271. [PMID: 22866075 DOI: 10.3892/ol.2010.228] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Accepted: 12/10/2010] [Indexed: 11/06/2022] Open
Abstract
7,12-Dimethylbenz(a)-anthracene (DMBA)-induced oral buccal mucosa squamous cell carcinoma in Syrian golden hamsters was used to establish precancerous lesions. Agilent rat whole-genome microarray and biological information analysis were used to screen for genes related to key diseases during the transformation of normal buccal mucosa to precancerous lesions in golden hamsters. DMBA acetone solution (0.5%) was used to establish a model of precancerous lesions in oral buccal mucosa in golden hamsters. The results showed that a total of 1331 genes were differentially expressed, including 1278 known, 53 unknown, 747 up-regulated and 584 down-regulated genes. Analysis revealed a total of 14 gene interaction pathways that significantly associated with the 1278 known differentially expressed genes (P<0.05). In conclusion, the occurrence of precancerous lesions in the oral buccal mucosa of golden hamsters was caused by a number of genetic changes that resulted in changes to their respective pathways. Key candidate genes for the formation of precancerous lesions in oral buccal mucosa included Cyp2b13, Orc1L, casp8, CCL5, CXCL12, CCL20, Serping1, P518/Qrfp, F5, TFPI, Vcam1, Fn1, Angpt2, Lcp2, Cxadr, Lyn, Hck, Btk, RGD1564385/fes, Vav1 and IL5ra.
Collapse
Affiliation(s)
- Dan Chen
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| | | | | | | | | |
Collapse
|
40
|
Wang X, Gao XH, Hong Y, Li X, Chen HD. Local hyperthermia decreases the expression of CCL-20 in condyloma acuminatum. Virol J 2010; 7:301. [PMID: 21050487 PMCID: PMC2991315 DOI: 10.1186/1743-422x-7-301] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Accepted: 11/04/2010] [Indexed: 12/02/2022] Open
Abstract
Background Local hyperthermia has been successfully used in the treatment of viral warts. However, the mechanism of action has largely remained unclear. CCL-20 (also known as MIP-3α) is the most potent chemokine for recruitment of Langerhans cell (LC) precursors into the skin. CCL-20 expression can be increased by TNF-α and IL-1α. The effects of local hyperthermia on the mRNA expressions of CCL-20, TNF-α, IL-1α have been investigated in both condyloma acuminata (CA) and normal skin. Under an organotypic culture condition, fresh CA and normal skin were subjected to surface heating at 37°C, 42°C and 45°C for 30 mins, respectively. Results The mRNA expressions of CCL-20 and IL-1α in CA specimen were significantly higher than those in normal skin. Local hyperthermia at 42°C and 45°C significantly decreased the mRNA levels of CCL-20 and IL-1α, as compared with the control groups (p < 0.01). The decrease of CCL-20 was well correlated with that of IL-1α. The expression of TNF-α in CA remained unchanged in spite of the temperature variation. Local hyperthermia at 45°C concomitantly increased the mRNA expression of CCL-20 and IL-1α in normal skin. Conclusions Our study suggests that hyperthermia decreases the expression of CCL-20 with concomitant decrease in IL-1α, and reduce the number of Langerhans cells in HPV infected skin.
Collapse
Affiliation(s)
- Xiaoqin Wang
- Department of Dermatology, No,1 Hospital of China Medical University, Shenyang 110001, PR China
| | | | | | | | | |
Collapse
|
41
|
Wang P, Yang X, Xu W, Li K, Chu Y, Xiong S. Integrating individual functional moieties of CXCL10 and CXCL11 into a novel chimeric chemokine leads to synergistic antitumor effects: a strategy for chemokine-based multi-target-directed cancer therapy. Cancer Immunol Immunother 2010; 59:1715-26. [PMID: 20706716 PMCID: PMC11030099 DOI: 10.1007/s00262-010-0901-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Accepted: 07/28/2010] [Indexed: 10/19/2022]
Abstract
The complexity of tumor biology necessitates a multimodality approach that targets different aspects of tumor environment in order to generate the greatest benefit. IFN-inducible T cell alpha chemoattractant (ITAC)/CXCL11 and IFN-inducible protein 10 (IP10)/CXCL10 could exert antitumor effects with functional specificity and thus emerge as attractive candidates for combinatorial strategy. Disappointedly, a synergistic antitumor effect could not be observed when CXCL10 and CXCL11 were pooled together. In this regard, we seek to improve antitumor efficacy by integrating their individual functional moieties into a chemokine chimeric molecule, designated ITIP, which was engineered by substituting the N-terminal and N-loop region of CXCL10 with those of CXCL11. The functional properties of ITIP were determined by chemotaxis and angiogenesis assays. The antitumor efficacy was tested in murine CT26 colon carcinoma, 4T1 mammary carcinoma and 3LL lung carcinoma. Here we showed that ITIP not only exhibited respective functional superiority but strikingly promoted regression of established tumors and remarkably prolonged survival of mice compared with its parent chemokines, either alone or in combination. The chemokine chimera induced an augmented anti-tumor immunity and a marked decrease in tumor vasculature. Antibody neutralization studies indicated that CXCL10 and CXCL11 moieties of ITIP were responsible for anti-angiogenesis and chemotaxis in antitumor response, respectively. These results indicated that integrating individual functional moieties of CXCL10 and CXCL11 into a chimeric chemokine could lead to a synergistic antitumor effect. Thus, this integration strategy holds promise for chemokine-based multiple targeted therapy of cancer.
Collapse
MESH Headings
- Animals
- Apoptosis
- Blotting, Western
- Calcium/metabolism
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/prevention & control
- Cell Adhesion
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Chemokine CXCL10/immunology
- Chemokine CXCL11/immunology
- Chemotaxis
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/prevention & control
- Drug Synergism
- Enzyme-Linked Immunosorbent Assay
- Female
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Fluorescent Antibody Technique
- Immunoenzyme Techniques
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Wound Healing
Collapse
Affiliation(s)
- Ping Wang
- Institute for Immunobiology, Department of Immunology, Shanghai Medical College, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Xiuli Yang
- Institute for Immunobiology, Department of Immunology, Shanghai Medical College, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Wei Xu
- Institute for Immunobiology, Department of Immunology, Shanghai Medical College, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Kang Li
- Institute for Immunobiology, Department of Immunology, Shanghai Medical College, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Yiwei Chu
- Institute for Immunobiology, Department of Immunology, Shanghai Medical College, Fudan University, Shanghai, 200032 People’s Republic of China
| | - Sidong Xiong
- Institute for Immunobiology, Department of Immunology, Shanghai Medical College, Fudan University, Shanghai, 200032 People’s Republic of China
- Institute of Biology and Medical Sciences, Soochow University, Suzhou, 215006 People’s Republic of China
| |
Collapse
|
42
|
Song EY, Shurin MR, Tourkova IL, Gutkin DW, Shurin GV. Epigenetic mechanisms of promigratory chemokine CXCL14 regulation in human prostate cancer cells. Cancer Res 2010; 70:4394-401. [PMID: 20460540 DOI: 10.1158/0008-5472.can-10-0427] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chemokines play the key role in initiating immune responses by regulating the attraction and homing of immune cells to the lymphoid and nonlymphoid tissues. CXCL14 is a chemokine that in tumors may act as chemoattractant for monocytes and dendritic cells (DC), which may modulate antitumor immune responses in certain cancers. In this study, we investigated the mechanisms of loss of CXCL14 in prostate cancer cells. Cell treatment with the demethylating agent 5-aza-2-deoxycytidine resulted in the recovery of CXCL14 mRNA and protein expression. Hypermethylated CpG island sequences encompassing the CXCL14 gene promoter were identified. The restoration of CXCL14 by 5-aza-2-deoxycytidine treatment had functional impact, based on the DC chemoattractant activity of conditioned medium from drug-treated cells. Conversely, CXCL14 removal from conditioned media by affinity chromatography abolished its chemotactic properties, confirming that functionally active CXCL14 was generated in prostate cancer cells by relieving its transcriptional silencing with 5-aza-2-deoxycytidine. Our findings offer the first direct evidence for epigenetic regulation of chemokine expression in tumor cells.
Collapse
Affiliation(s)
- Eun Young Song
- Seoul National University College of Medicine, Seoul, South Korea
| | | | | | | | | |
Collapse
|
43
|
Intratumoral DNA electroporation induces anti-tumor immunity and tumor regression. Cancer Immunol Immunother 2010; 59:409-17. [PMID: 19730859 DOI: 10.1007/s00262-009-0760-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 08/21/2009] [Indexed: 12/27/2022]
Abstract
In situ expression of a foreign antigen and an immune-modulating cytokine by intratumoral DNA electroporation was tested as a cancer therapy regimen. Transgene expression in the tumors was sustained for 2-3 weeks after intratumoral electroporation with mammalian expression plasmid containing firefly luciferase cDNA. Electroporation with cDNA encoding tetanus toxin fragment C (TetC) induced tetanus toxin-binding antibody, demonstrating immune recognition of the transgene product. Intratumoral electroporation with TetC and IL-12 cDNA after mice were treated with CD25 mAb to remove regulatory T cells induced IFN-gamma producing T-cell response to tumor-associated antigen, heavy inflammatory infiltration, regression of established tumors and immune memory to protect mice from repeated tumor challenge. Intratumoral expression of immune-modulating molecules may be most suitable in the neoadjuvant setting to enhance the therapeutic efficacy and provide long-term protection.
Collapse
|
44
|
Abstract
Chemokines are a key component of cancer-related inflammation. Chemokines and chemokine receptors are downstream of genetic events that cause neoplastic transformation and are components of chronic inflammatory conditions, which predispose to cancer. Components of the chemokine system affect in a cell autonomous or non-autonomous way multiple pathways of tumor progression, including: leukocyte recruitment and function; cellular senescence; tumor cell proliferation and survival; invasion and metastasis. Available information in preclinical and clinical settings suggests that the chemokine system represents a valuable target for the development of innovative therapeutic strategies.
Collapse
|
45
|
Jassem W, Fuggle S, Thompson R, Arno M, Taylor J, Byrne J, Heaton N, Rela M. Effect of ischemic preconditioning on the genomic response to reperfusion injury in deceased donor liver transplantation. Liver Transpl 2009; 15:1750-65. [PMID: 19938126 DOI: 10.1002/lt.21936] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ischemic preconditioning (IP) is an effective method for protecting organs from ischemia/reperfusion (IR) injury; however, the molecular basis of this protective effect is poorly understood. This study assessed the gene expression profile in liver allografts during transplantation and evaluated the impact of IP. Prereperfusion and postreperfusion biopsy specimens from livers subjected to IP (n = 19) or no preconditioning (the IR group; n = 16) were obtained. Total RNA was extracted and hybridized to GeneChip microarrays, and the findings were validated with real-time quantitative reverse-transcription polymerase chain reaction (qRT-PCR). IP livers showed less of an increase in aspartate aminotransferase after transplantation. A microarray analysis of the IR group showed increased expression of 57 genes mainly involved in cell death, inflammation and immune response, stress, and modulation of the cell cycle. The IP group showed attenuation of the expression of these genes after reperfusion. Additionally, IP led to increased expression of 43 genes involved in growth and maintenance, cell-cycle regulation, proliferation, and development. The expression of the 12 most significant genes was validated in all patients with real-time qRT-PCR, and the fold changes of a number of genes correlated with clinical parameters and graft outcomes. IP protection of liver allografts was associated with a reduction in the expression of immune response genes and promotion of those involved in protection and repair.
Collapse
Affiliation(s)
- Wayel Jassem
- Liver Transplant Unit, Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Sengupta N, MacFie TS, MacDonald TT, Pennington D, Silver AR. Cancer immunoediting and "spontaneous" tumor regression. Pathol Res Pract 2009; 206:1-8. [PMID: 19945228 DOI: 10.1016/j.prp.2009.10.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Accepted: 10/07/2009] [Indexed: 12/28/2022]
Abstract
The combination of host protective and tumor-promoting actions of the immune system throughout tumor development is termed cancer immunoediting. This review briefly summarizes the currently vast evidence supporting the immune system's role in not only protecting against developing cancer, but also sculpting tumor immunogenicity and immune escape. We also briefly summarize the history of immunotherapy and discuss the immunoediting process in the context of spontaneous tumor regression and whether this observation can be utilized in future treatment regimens.
Collapse
Affiliation(s)
- Neel Sengupta
- Colorectal Cancer Genetics, Centre for Academic Surgery, Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, Whitechapel, London E1 2AT, UK
| | | | | | | | | |
Collapse
|
47
|
Wang YS, Li D, Shi HS, Wen YJ, Yang L, Xu N, Chen XC, Chen X, Chen P, Li J, Deng HX, Wang CT, Xie G, Huang S, Mao YQ, Chen LJ, Zhao X, Wei YQ. Intratumoral expression of mature human neutrophil peptide-1 mediates antitumor immunity in mice. Clin Cancer Res 2009; 15:6901-11. [PMID: 19861439 DOI: 10.1158/1078-0432.ccr-09-0484] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE Human neutrophil peptides (HNP1-3), small molecular antimicrobial peptides, are expressed within tumors and associated with tumor necrosis and inhibition of angiogenesis. Recent investigations have suggested that HNP1-3 are likely to be involved in the host immune responses to tumors. EXPERIMENTAL DESIGN We used recombinant pSec-HNP1, which expresses a secretable form of HNP1, to obtain expression of HNP1 in the tumor milieu in immunocompetent mice to explore the possible roles of HNP1 in tumor immunity. The antitumor effects were investigated in established CT26 colon cancer and 4T1 breast cancer models. RESULTS HNP1-mediated chemotactic and activating effects on immature dendritic cells were detected both in vitro and in vivo. Intratumoral expression of HNP1 resulted in not only significant tumor growth inhibition but also increased CTL infiltration within tumors. Adoptive transfer of splenocytes and a (51)Cr release assay revealed specific cellular immune responses. Furthermore, increased antibodies were also found in sera from pSec-HNP1-treated mice supporting specific humoral immune responses. Increased apoptosis and decreased angiogenesis were also shown in treated tumors. CONCLUSIONS These findings indicate that HNP1 can exert multiple antitumor effects through different mechanisms; more importantly, HNP1 mediates host immune responses to tumors in situ through the recruitment and subsequent activation of immature dendritic cells and thus shows promising potential in cancer therapy.
Collapse
Affiliation(s)
- Yong-Sheng Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Department of Gynecology and Obstetrics, Second West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Franciszkiewicz K, Le Floc'h A, Jalil A, Vigant F, Robert T, Vergnon I, Mackiewicz A, Benihoud K, Validire P, Chouaib S, Combadière C, Mami-Chouaib F. Intratumoral Induction of CD103 Triggers Tumor-Specific CTL Function and CCR5-Dependent T-Cell Retention. Cancer Res 2009; 69:6249-55. [DOI: 10.1158/0008-5472.can-08-3571] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
Conditioning vaccination site with irradiated MIP-3alpha-transfected tumor cells enhances efficacy of dendritic cell-based cancer vaccine. J Immunother 2009; 32:363-9. [PMID: 19342969 DOI: 10.1097/cji.0b013e31819d29d8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Macrophage inflammation protein-3alpha (MIP-3alpha) is a chemokine expressed in inflamed tissue and capable of inducing migration of immature dendritic cells (DCs) or Langerhans cells. We postulated that conditioning vaccination sites with MIP-3alpha might enhance the efficacy of subsequently administered DC-based cancer vaccines. Our results demonstrate that subcutaneously injection of irradiated tumor cells expressing MIP-3alpha induces substantial cell infiltration to the injection site. Vaccination of irradiated tumor cells expressing MIP-3alpha followed by DCs pulsed with irradiated tumor cells can effectively suppress tumor growth in animals, which is significantly better than vaccination with irradiated MIP-3alpha-producing tumor cells or DCs pulsed with tumor cells alone. The protective effect was most evident when the MIP-3alpha-producing tumor cells and DC-based vaccines were injected at the same site. These results support the notion that this combination vaccination strategy might generate a more effective immune response to suppress the growth of tumor cells in animals.
Collapse
|
50
|
THORBURN NEE KRASNA ELISKA, KOLESAR LIBOR, BRABCOVA EVA, PETRICKOVA KATERINA, PETRICEK MIROSLAV, JARESOVA MARCELA, SLAVCEV ANTONIJ, STRIZ ILJA. CXC and CC chemokines induced in human renal epithelial cells by inflammatory cytokines. APMIS 2009; 117:477-87. [DOI: 10.1111/j.1600-0463.2009.02446.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|