1
|
Halliez C, Ibrahim H, Otonkoski T, Mallone R. In vitro beta-cell killing models using immune cells and human pluripotent stem cell-derived islets: Challenges and opportunities. Front Endocrinol (Lausanne) 2023; 13:1076683. [PMID: 36726462 PMCID: PMC9885197 DOI: 10.3389/fendo.2022.1076683] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Type 1 diabetes (T1D) is a disease of both autoimmunity and β-cells. The β-cells play an active role in their own demise by mounting defense mechanisms that are insufficient at best, and that can become even deleterious in the long term. This complex crosstalk is important to understanding the physiological defense mechanisms at play in healthy conditions, their alterations in the T1D setting, and therapeutic agents that may boost such mechanisms. Robust protocols to develop stem-cell-derived islets (SC-islets) from human pluripotent stem cells (hPSCs), and islet-reactive cytotoxic CD8+ T-cells from peripheral blood mononuclear cells offer unprecedented opportunities to study this crosstalk. Challenges to develop in vitro β-cell killing models include the cluster morphology of SC-islets, the relatively weak cytotoxicity of most autoimmune T-cells and the variable behavior of in vitro expanded CD8+ T-cells. These challenges may however be highly rewarding in light of the opportunities offered by such models. Herein, we discuss these opportunities including: the β-cell/immune crosstalk in an islet microenvironment; the features that make β-cells more sensitive to autoimmunity; therapeutic agents that may modulate β-cell vulnerability; and the possibility to perform analyses in an autologous setting, i.e., by generating T-cell effectors and SC-islets from the same donor.
Collapse
Affiliation(s)
- Clémentine Halliez
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| | - Hazem Ibrahim
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
- Department of Pediatrics, Helsinki University Hospital, Helsinki, Finland
| | - Roberto Mallone
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| |
Collapse
|
2
|
Dada S, Ellis SLS, Wood C, Nohara LL, Dreier C, Garcia NH, Saranchova I, Munro L, Pfeifer CG, Eyford BA, Kari S, Garrovillas E, Caspani G, Al Haddad E, Gray PW, Morova T, Lack NA, Andersen RJ, Tjoelker L, Jefferies WA. Specific cannabinoids revive adaptive immunity by reversing immune evasion mechanisms in metastatic tumours. Front Immunol 2022; 13:982082. [PMID: 36923728 PMCID: PMC10010394 DOI: 10.3389/fimmu.2022.982082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/20/2022] [Indexed: 02/24/2023] Open
Abstract
Emerging cancers are sculpted by neo-Darwinian selection for superior growth and survival but minimal immunogenicity; consequently, metastatic cancers often evolve common genetic and epigenetic signatures to elude immune surveillance. Immune subversion by metastatic tumours can be achieved through several mechanisms; one of the most frequently observed involves the loss of expression or mutation of genes composing the MHC-I antigen presentation machinery (APM) that yields tumours invisible to Cytotoxic T lymphocytes, the key component of the adaptive cellular immune response. Fascinating ethnographic and experimental findings indicate that cannabinoids inhibit the growth and progression of several categories of cancer; however, the mechanisms underlying these observations remain clouded in uncertainty. Here, we screened a library of cannabinoid compounds and found molecular selectivity amongst specific cannabinoids, where related molecules such as Δ9-tetrahydrocannabinol, cannabidiol, and cannabigerol can reverse the metastatic immune escape phenotype in vitro by inducing MHC-I cell surface expression in a wide variety of metastatic tumours that subsequently sensitizing tumours to T lymphocyte recognition. Remarkably, H3K27Ac ChIPseq analysis established that cannabigerol and gamma interferon induce overlapping epigenetic signatures and key gene pathways in metastatic tumours related to cellular senescence, as well as APM genes involved in revealing metastatic tumours to the adaptive immune response. Overall, the data suggest that specific cannabinoids may have utility in cancer immunotherapy regimens by overcoming immune escape and augmenting cancer immune surveillance in metastatic disease. Finally, the fundamental discovery of the ability of cannabinoids to alter epigenetic programs may help elucidate many of the pleiotropic medicinal effects of cannabinoids on human physiology.
Collapse
Affiliation(s)
- Sarah Dada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Samantha L S Ellis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Christi Wood
- Biotechnology - Biomedical Science and Technology (BST), University of Applied Sciences, Mannheim, Germany
| | - Lilian L Nohara
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Carola Dreier
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.,Biotechnology - Biomedical Science and Technology (BST), University of Applied Sciences, Mannheim, Germany
| | | | - Iryna Saranchova
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Lonna Munro
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Cheryl G Pfeifer
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Brett A Eyford
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Suresh Kari
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Emmanuel Garrovillas
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Giorgia Caspani
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Eliana Al Haddad
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | | | - Tunc Morova
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Nathan A Lack
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,School of Medicine, Koç University, Istanbul, Türkiye
| | - Raymond J Andersen
- Department of Chemistry, University of British Columbia, Vancouver, BC, Canada
| | | | - Wilfred A Jefferies
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,Department of Zoology, University of British Columbia, Vancouver, BC, Canada.,Department of Urological Science, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
3
|
A Radioactive-Free Method for the Thorough Analysis of the Kinetics of Cell Cytotoxicity. J Imaging 2021; 7:jimaging7110222. [PMID: 34821853 PMCID: PMC8624610 DOI: 10.3390/jimaging7110222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 11/17/2022] Open
Abstract
The cytotoxic activity of T cells and Natural Killer cells is usually measured with the chromium release assay (CRA), which involves the use of 51Chromium (51Cr), a radioactive substance dangerous to the operator and expensive to handle and dismiss. The accuracy of the measurements depends on how well the target cells incorporate 51Cr during labelling which, in turn, depends on cellular division. Due to bystander metabolism, the target cells spontaneously release 51Cr, producing a high background noise. Alternative radioactive-free methods have been developed. Here, we compare a bioluminescence (BLI)-based and a carboxyfluorescein succinimidyl ester (CFSE)-based cytotoxicity assay to the standard radioactive CRA. In the first assay, the target cells stably express the enzyme luciferase, and vitality is measured by photon emission upon the addition of the substrate d-luciferin. In the second one, the target cells are labelled with CFSE, and the signal is detected by Flow Cytometry. We used these two protocols to measure cytotoxicity induced by treatment with NK cells. The cytotoxicity of NK cells was determined by adding increasing doses of human NK cells. The results obtained with the BLI method were consistent with those obtained with the CRA- or CFSE-based assays 4 hours after adding the NK cells. Most importantly, with the BLI assay, the kinetic of NK cells’ killing was thoroughly traced with multiple time point measurements, in contrast with the single time point measurement the other two methods allow, which unveiled additional information on NK cell killing pathways.
Collapse
|
4
|
Wang Z, Ran L, Chen C, Shi R, Dong Y, Li Y, Zhou X, Qi Y, Zhu P, Gao Y, Wu Y. Identification of HLA-A2-Restricted Mutant Epitopes from Neoantigens of Esophageal Squamous Cell Carcinoma. Vaccines (Basel) 2021; 9:vaccines9101118. [PMID: 34696226 PMCID: PMC8541546 DOI: 10.3390/vaccines9101118] [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: 08/07/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/24/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC), one of the deadliest gastrointestinal cancers, has had limited effective therapeutic strategies up to now. Accumulating evidence suggests that effective immunotherapy in cancer patients has been associated with T cells responsive to mutant peptides derived from neoantigens. Here, we selected 35 human leukocyte antigen-A2 (HLA-A2)-restricted mutant (MUT) peptides stemmed from neoantigens of ESCC. Among them, seven mutant peptides had potent binding affinity to HLA-A*0201 molecules and could form a stable peptide/HLA-A*0201 complex. Three mutant peptides (TP53-R267P, NFE2L2-D13N, and PCLO-E4090Q) of those were immunogenic and could induce the cytotoxic T lymphocytes (CTLs) recognizing mutant peptides presented on transfected cells in an HLA-A2-restricted and MUT peptide-specific manner. In addition, the CTL response in immunized HLA-A2.1/Kb transgenic (Tg) mice was enhanced by coupling MUT peptides to peptide WH, a peptide delivery carrier targeting Clec9a+ DCs. Then, the possible binding model conversions between the WT and MUT candidate peptides were analyzed by docking with the pockets of HLA-A*0201 molecule. We therefore propose a novel strategy and epitopes for immunotherapy of ESCC based on neoantigens.
Collapse
Affiliation(s)
- Zhiwei Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (L.R.); (C.C.); (R.S.); (Y.D.); (Y.L.); (X.Z.); (Y.Q.); (P.Z.)
| | - Ling Ran
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (L.R.); (C.C.); (R.S.); (Y.D.); (Y.L.); (X.Z.); (Y.Q.); (P.Z.)
| | - Chunxia Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (L.R.); (C.C.); (R.S.); (Y.D.); (Y.L.); (X.Z.); (Y.Q.); (P.Z.)
| | - Ranran Shi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (L.R.); (C.C.); (R.S.); (Y.D.); (Y.L.); (X.Z.); (Y.Q.); (P.Z.)
| | - Yu Dong
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (L.R.); (C.C.); (R.S.); (Y.D.); (Y.L.); (X.Z.); (Y.Q.); (P.Z.)
| | - Yubing Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (L.R.); (C.C.); (R.S.); (Y.D.); (Y.L.); (X.Z.); (Y.Q.); (P.Z.)
| | - Xiuman Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (L.R.); (C.C.); (R.S.); (Y.D.); (Y.L.); (X.Z.); (Y.Q.); (P.Z.)
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (L.R.); (C.C.); (R.S.); (Y.D.); (Y.L.); (X.Z.); (Y.Q.); (P.Z.)
- International Joint Laboratory for Protein and Peptide Drugs of Henan Province, Zhengzhou University, Zhengzhou 450001, China
| | - Pingping Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (L.R.); (C.C.); (R.S.); (Y.D.); (Y.L.); (X.Z.); (Y.Q.); (P.Z.)
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 510080, China
- Correspondence: (Y.G.); (Y.W.); Tel./Fax: +86-371-6778-3235 (Y.W.)
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (L.R.); (C.C.); (R.S.); (Y.D.); (Y.L.); (X.Z.); (Y.Q.); (P.Z.)
- International Joint Laboratory for Protein and Peptide Drugs of Henan Province, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: (Y.G.); (Y.W.); Tel./Fax: +86-371-6778-3235 (Y.W.)
| |
Collapse
|
5
|
Shi R, Li Y, Ran L, Dong Y, Zhou X, Tang J, Han L, Wang M, Pang L, Qi Y, Wu Y, Gao Y. Screening and identification of HLA-A2-restricted neoepitopes for immunotherapy of non-microsatellite instability-high colorectal cancer. SCIENCE CHINA-LIFE SCIENCES 2021; 65:572-587. [PMID: 34236583 DOI: 10.1007/s11427-021-1944-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/12/2021] [Indexed: 12/27/2022]
Abstract
Colorectal cancer has one of the highest mortality rates among malignant tumors, and most patients with non-microsatellite instability-high (MSI-H) colorectal cancer do not benefit from targeted therapy or immune checkpoint inhibitors. Identification of immunogenic neoantigens is a promising strategy for inducing specific antitumor T cells for cancer immunotherapy. Here, we screened potential high-frequency neoepitopes from non-MSI-H colorectal cancer and tested their abilities to induce tumor-specific cytotoxic T cell responses. Three HLA-A2-restricted neoepitopes (P31, P50, and P52) were immunogenic and could induce cytotoxic T lymphocytes in peripheral blood mononuclear cells from healthy donors and colorectal cancer patients. Cytotoxic T lymphocytes induced in HLA-A2.1/Kb transgenic mice could recognize and lyse mutant neoepitope-transfected HLA-A2+ cancer cells. Adoptive transfer of cytotoxic T lymphocytes induced by the peptide pool of these three neoepitopes effectively inhibited tumor growth and increased the therapeutic effects of anti-PD-1 antibody. These results revealed the potential of high-frequency mutation-specific peptide-based immunotherapy as a personalized treatment approach for patients with non-MSI-H colorectal cancer. The combination of adoptive T cell therapy based on these neoepitopes with immune checkpoint inhibitors, such as anti-PD-1, could provide a promising treatment strategy for non-MSI-H colorectal cancer.
Collapse
Affiliation(s)
- Ranran Shi
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yubing Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Ling Ran
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yu Dong
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiuman Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jingwen Tang
- Department of Integrated Chinse and Western Medicine, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Lu Han
- Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Mingshuang Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Liwei Pang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Bioactive Macromolecules, Zhengzhou University, Zhengzhou, 450001, China
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Bioactive Macromolecules, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yanfeng Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
6
|
Chen J, Wu Y, Duan FX, Wang SN, Guo XY, Ding SQ, Zhou JH, Hu JG, Lü HZ. Effect of M2 macrophage adoptive transfer on transcriptome profile of injured spinal cords in rats. Exp Biol Med (Maywood) 2019; 244:880-892. [PMID: 31159561 DOI: 10.1177/1535370219854668] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The previous studies showed that alternatively activated anti-inflammatory macrophage (M2) adoptive immunity can improve the proportion of local M2 cells and play the neuroprotective effect after spinal cord injury (SCI). Its molecular mechanism is not yet very clear. Therefore, this study aims to analyze the effect of the M2 adoptive transfer on the local expression of gene transcription. Sprague-Dawley (SD) rats were used for culture of macrophages and establishment of SCI models. After SCI, the polarized M2 macrophages were transferred to the injured rats by tail vein injection. Seven days after operation, the differentially expressed genes (DEGs) in the spinal cords were analyzed by RNA-sequencing (RNA-Seq). Then, the functional enrichment analysis and pathways were performed by using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), respectively. RNA-Seq showed that M2 adoptive immunity can down-regulate many well-studied gene expressions associated with signaling pathways of inflammatory, such as antigen processing and presentation, phagosome, cell adhesion molecules, natural killer cell-mediated cytotoxicity, endocytosis, proteasome, and Toll-like receptor signaling pathway. These may explain the mechanism of our previous adoptive immunization of M2 cells to provide neuroprotection for SCI. In addition, a novel pathway, retinoic acid-inducible gene-1 (RIG-I)-like receptor signaling pathway was found to be involved in the pathological process of SCI and the response to M2 adoptive immunity as well. This will provide a new explanation for the pathological mechanism of SCI and a new theoretical and experimental basis for its clinical treatment. The raw Illumina data are available at http://www.ncbi.nlm.nih.gov/sra (accession number PRJNA517238). Impact statement This research aimed to analyze the effect of M2 macrophage adoptive transfer on the local expression of gene transcription after SCI by RNA-Seq. The results showed that M2 adoptive immunity can down-regulate many well-studied gene expressions associated with signaling pathways of inflammatory. These may explain the mechanism of our previous adoptive immunization of M2 cells to provide neuroprotection for SCI. In addition, a novel pathway, RIG-I-like receptor signaling pathway was also found to involve in the pathological process of SCI and the response to M2 adoptive immunity. This will provide a new explanation for the pathological mechanism of SCI and a new theoretical and experimental basis for its clinical treatment.
Collapse
Affiliation(s)
- Jing Chen
- 1 Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,2 Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,3 Department of Immunology, Bengbu Medical College, Anhui 233030, PR China
| | - Yan Wu
- 1 Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,2 Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,3 Department of Immunology, Bengbu Medical College, Anhui 233030, PR China
| | - Fei-Xiang Duan
- 1 Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,2 Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Sai-Nan Wang
- 1 Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,2 Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,3 Department of Immunology, Bengbu Medical College, Anhui 233030, PR China
| | - Xue-Yan Guo
- 1 Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,2 Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Shu-Qin Ding
- 1 Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,2 Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Ji-Hong Zhou
- 3 Department of Immunology, Bengbu Medical College, Anhui 233030, PR China
| | - Jian-Guo Hu
- 1 Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,2 Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - He-Zuo Lü
- 1 Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,2 Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China.,3 Department of Immunology, Bengbu Medical College, Anhui 233030, PR China
| |
Collapse
|
7
|
de Wolf C, van de Bovenkamp M, Hoefnagel M. Regulatory perspective on in vitro potency assays for human T cells used in anti-tumor immunotherapy. Cytotherapy 2018; 20:601-622. [PMID: 29598903 DOI: 10.1016/j.jcyt.2018.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/25/2018] [Accepted: 01/27/2018] [Indexed: 02/06/2023]
Abstract
The adaptive immune system is known to play an important role in anti-neoplastic responses via induction of several effector pathways, resulting in tumor cell death. Because of their ability to specifically recognize and kill tumor cells, the potential use of autologous tumor-derived and genetically engineered T cells as adoptive immunotherapy for cancer is currently being explored. Because of the variety of potential T cell-based medicinal products at the level of starting material and manufacturing process, product-specific functionality assays are needed to ensure quality for individual products. In this review, we provide an overview of in vitro potency assays suggested for characterization and release of different T cell-based anti-tumor products. We discuss functional assays, as presented in scientific advices and literature, highlighting specific advantages and limitations of the various assays. Because the anticipated in vivo mechanism of action for anti-tumor T cells involves tumor recognition and cell death, in vitro potency assays based on the cytotoxic potential of antigen-specific T cells are most evident. However, assays based on other T cell properties may be appropriate as surrogates for cytotoxicity. For all proposed assays, biological relevance of the tests and correlation of the read-outs with in vivo functionality need to be substantiated with sufficient product-specific (non-)clinical data. Moreover, further unraveling the complex interaction of immune cells with and within the tumor environment is expected to lead to further improvement of the T cell-based products. Consequently, increased knowledge will allow further optimized guidance for potency assay development.
Collapse
Affiliation(s)
- Charlotte de Wolf
- Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands; Department of Infectious Diseases and Immunology, Utrecht University, The Netherlands
| | | | | |
Collapse
|
8
|
Wonderlich J, Shearer G, Livingstone A, Brooks A, Soloski MJ, Presby MM. Induction and Measurement of Cytotoxic T Lymphocyte Activity. ACTA ACUST UNITED AC 2018; 120:3.11.1-3.11.29. [PMID: 29512145 DOI: 10.1002/cpim.38] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cytotoxic T cells (CTLs) are important immune effector cells in the adaptive immune response. It has been well documented that CTLs are important in host immune responses to viral and bacterial intracellular pathogens, tumors, and transplanted tissues. The properties of CTLs have been studied extensively in murine models, and their roles validated in the human setting. Frequently, the presence of these cells correlates well with protective immunity, so the ability to readily measure the activity of these cells is an important immunological measurement. In this unit, several assays are described that are commonly utilized to induce CTLs and to measure CTL activity both in vitro and in vivo. These assays are adaptable to many experimental and/or disease models, and in the case of the in vitro assays can be applied to measure CTL activity in human samples. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
| | | | | | - Andrew Brooks
- University of Melbourne, Victoria, Australia (CTL activity in vivo)
| | - Mark J Soloski
- Johns Hopkins School of Medicine, Baltimore, Maryland (non-radioactive assay for in vitro CTL activity)
| | - Matthew M Presby
- Johns Hopkins School of Medicine, Baltimore, Maryland (non-radioactive assay for in vitro CTL activity)
| |
Collapse
|
9
|
Chen Y, Xue SA, Behboudi S, Mohammad GH, Pereira SP, Morris EC. Ex Vivo PD-L1/PD-1 Pathway Blockade Reverses Dysfunction of Circulating CEA-Specific T Cells in Pancreatic Cancer Patients. Clin Cancer Res 2017; 23:6178-6189. [PMID: 28710313 PMCID: PMC5683391 DOI: 10.1158/1078-0432.ccr-17-1185] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/20/2017] [Accepted: 07/10/2017] [Indexed: 12/27/2022]
Abstract
Purpose: Carcinoembryonic antigen (CEA) is a candidate target for cellular immunotherapy of pancreatic cancer. In this study, we have characterized the antigen-specific function of autologous cytotoxic T lymphocytes (CTL) specific for the HLA-A2-restricted peptide, pCEA691-699, isolated from the peripheral T-cell repertoire of pancreatic cancer patients and sought to determine if ex vivo PD-L1 and TIM-3 blockade could enhance CTL function.Experimental Design: CD8+ T-cell lines were generated from peripheral blood mononuclear cells of 18 HLA-A2+ patients with pancreatic cancer and from 15 healthy controls. In vitro peptide-specific responses were evaluated by flow cytometry after staining for intracellular cytokine production and carboxy fluorescein succinimydyl ester cytotoxicity assays using pancreatic cancer cell lines as targets.Results: Cytokine-secreting functional CEA691-specific CTL lines were successfully generated from 10 of 18 pancreatic cancer patients, with two CTL lines able to recognize and kill both CEA691 peptide-loaded T2 cells and CEA+ HLA-A2+ pancreatic cancer cell lines. In the presence of ex vivo PD-L1 blockade, functional CEA691-specific CD8+ T-cell responses, including IFNγ secretion and proliferation, were enhanced, and this effect was more pronounced on Ag-specific T cells isolated from tumor draining lymph nodes.Conclusions: These data demonstrate that CEA691-specific CTL can be readily expanded from the self-restricted T-cell repertoire of pancreatic cancer patients and that their function can be enhanced by PD-L1 blockade. Clin Cancer Res; 23(20); 6178-89. ©2017 AACR.
Collapse
Affiliation(s)
- Yuan Chen
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Shao-An Xue
- Institute of Immunity and Transplantation, University College London, London, United Kingdom.,Genetic Engineering Laboratory, School of Biological and Environmental Engineering, Xi'An University, Xi'An, P. R. China
| | | | - Goran H Mohammad
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,Chemistry Department, College of Science, University of Sulaimani, Sulaimanyah, Kurdistan Region, Iraq
| | - Stephen P Pereira
- Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Emma C Morris
- Institute of Immunity and Transplantation, University College London, London, United Kingdom.
| |
Collapse
|
10
|
Holgado MP, Falivene J, Maeto C, Amigo M, Pascutti MF, Vecchione MB, Bruttomesso A, Calamante G, Del Médico-Zajac MP, Gherardi MM. Deletion of A44L, A46R and C12L Vaccinia Virus Genes from the MVA Genome Improved the Vector Immunogenicity by Modifying the Innate Immune Response Generating Enhanced and Optimized Specific T-Cell Responses. Viruses 2016; 8:E139. [PMID: 27223301 PMCID: PMC4885094 DOI: 10.3390/v8050139] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/02/2016] [Accepted: 05/11/2016] [Indexed: 12/31/2022] Open
Abstract
MVA is an attenuated vector that still retains immunomodulatory genes. We have previously reported its optimization after deleting the C12L gene, coding for the IL-18 binding-protein. Here, we analyzed the immunogenicity of MVA vectors harboring the simultaneous deletion of A44L, related to steroid synthesis and A46R, a TLR-signaling inhibitor (MVAΔA44L-A46R); or also including a deletion of C12L (MVAΔC12L/ΔA44L-A46R). The absence of biological activities of the deleted genes in the MVA vectors was demonstrated. Adaptive T-cell responses against VACV epitopes, evaluated in spleen and draining lymph-nodes of C57Bl/6 mice at acute/memory phases, were of higher magnitude in those animals that received deleted MVAs compared to MVAwt. MVAΔC12L/ΔA44L-A46R generated cellular specific memory responses of higher quality characterized by bifunctionality (CD107a/b⁺/IFN-γ⁺) and proliferation capacity. Deletion of selected genes from MVA generated innate immune responses with higher levels of determining cytokines related to T-cell response generation, such as IL-12, IFN-γ, as well as IL-1β and IFN-β. This study describes for the first time that simultaneous deletion of the A44L, A46R and C12L genes from MVA improved its immunogenicity by enhancing the host adaptive and innate immune responses, suggesting that this approach comprises an appropriate strategy to increase the MVA vaccine potential.
Collapse
Affiliation(s)
- María Pía Holgado
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Facultad de Medicina, Ciudad de Buenos Aires 1121, Argentina.
| | - Juliana Falivene
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Facultad de Medicina, Ciudad de Buenos Aires 1121, Argentina.
| | - Cynthia Maeto
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Facultad de Medicina, Ciudad de Buenos Aires 1121, Argentina.
| | - Micaela Amigo
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Facultad de Medicina, Ciudad de Buenos Aires 1121, Argentina.
| | - María Fernanda Pascutti
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Facultad de Medicina, Ciudad de Buenos Aires 1121, Argentina.
| | - María Belén Vecchione
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Facultad de Medicina, Ciudad de Buenos Aires 1121, Argentina.
- Unidad de Microanálisis y Métodos Físicos Aplicados a Química Orgánica (UMYMFOR), Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires 1428, Argentina.
| | - Andrea Bruttomesso
- Unidad de Microanálisis y Métodos Físicos Aplicados a Química Orgánica (UMYMFOR), Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires 1428, Argentina.
| | - Gabriela Calamante
- Instituto de Biotecnología, CICVyA-INTA Castelar, Buenos Aires 1686, Argentina.
| | | | - María Magdalena Gherardi
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires-CONICET, Facultad de Medicina, Ciudad de Buenos Aires 1121, Argentina.
| |
Collapse
|
11
|
Delavari S, Sohrabi M, Ardestani MS, Faezi S, Tebianian M, Taghizadeh M, Shajiei A, Hosseini SY, Moghaddampour M, Mahdavi M. Pseudomonas aeruginosa flagellin as an adjuvant: superiority of a conjugated form of flagellin versus a mixture with a human immunodeficiency virus type 1 vaccine candidate in the induction of immune responses. J Med Microbiol 2015; 64:1361-1368. [PMID: 26404477 DOI: 10.1099/jmm.0.000174] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In the present study, the adjuvant activity of flagellin was compared, in the conjugated and mixed forms, against a peptide vaccine from human immunodeficiency virus type 1 (HIV-1) p24-Nef vaccine candidate. Mice were immunized with the HIV-1 p24-Nef peptide with flagellin in both conjugated and mixed forms. Lymphocyte proliferation, CTL activity, and IL-4 and IFN-γ cytokines were evaluated by bromodeoxyuridine, carboxyfluorescein succinimidyl ester and ELISA methods, respectively. At the same time, the frequency of IFN-γ-producing T-lymphocytes, as well as total and isotype-specific antibodies, were assessed by ELISPOT and indirect ELISA, respectively. Our experimental results showed that the immunized mice with the HIV-1 p24-Nef conjugated or mixed forms of flagellin led to increases in the proliferative responses and Th1 cytokine pattern. The conjugated form of vaccine led to increased CTL activity and a Th1 cytokine pattern of immune responses, as well as an IgM isotype of humoral responses in comparison with the mixed form. The flagellin-conjugated vaccine seems to be more potent in increasing vaccine immunogenicity.
Collapse
Affiliation(s)
- Safura Delavari
- Department of Biology, Faculty of Sciences, Qom branch of Islamic Azad University, Qom, Iran
| | - Mojtaba Sohrabi
- Department of Biology, Faculty of Sciences, Qom branch of Islamic Azad University, Qom, Iran
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sobhan Faezi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Majid Tebianian
- Department of Biotechnology, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Morteza Taghizadeh
- Department of Medical Virology, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Arezoo Shajiei
- Molecular Pathology Laboratory, Cancer Molecular Pathology Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Moghaddampour
- Department of Medical Virology, Razi Vaccine and Serum Research Institute, Karaj, Iran.,Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mahdavi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
12
|
Ito H, Ando T, Arioka Y, Saito K, Seishima M. Inhibition of indoleamine 2,3-dioxygenase activity enhances the anti-tumour effects of a Toll-like receptor 7 agonist in an established cancer model. Immunology 2015; 144:621-30. [PMID: 25322876 DOI: 10.1111/imm.12413] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/02/2014] [Accepted: 10/14/2014] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptor (TLR) agonists have been shown to have anti-tumour activity in basic research and clinical studies. However, TLR agonist monotherapy does not sufficiently eliminate tumours. Activation of the innate immune response by TLR agonists is effective at driving adaptive immunity via interleukin-12 (IL-12) or IL-1, but is counteracted by the simultaneous induction of immunosuppressive cytokines and other molecules, including IL-10, transforming growth factor-β, and indoleamine 2,3-dioxygenase (IDO). In the present study, we evaluated the anti-cancer effect of the TLR7 agonist, imiquimod (IMQ), in the absence of IDO activity. The administration of IMQ in IDO knockout (KO) mice inoculated with tumour cells significantly suppressed tumour progression compared with that in wild-type (WT) mice, and improved the survival rate. Moreover, injection with IMQ enhanced the tumour antigen-specific T helper type 1 response in IDO-KO mice with tumours. Combination therapy with IMQ and an IDO inhibitor also significantly inhibited tumour growth. Our results indicated that the enhancement of IDO expression with TLR agonists in cancer treatment might impair host anti-tumour immunity while the inhibition of IDO could enhance the therapeutic efficacy of TLR agonists via the increase of T helper type 1 immune response.
Collapse
Affiliation(s)
- Hiroyasu Ito
- Department of Informative Clinical Medicine, First Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | | | | | | | | |
Collapse
|
13
|
Ando T, Ito H, Arioka Y, Ogiso H, Seishima M. Combination therapy with α-galactosylceramide and a Toll-like receptor agonist exerts an augmented suppressive effect on lung tumor metastasis in a mouse model. Oncol Rep 2014; 33:826-32. [PMID: 25434827 DOI: 10.3892/or.2014.3634] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 11/04/2014] [Indexed: 11/06/2022] Open
Abstract
α-galactosylceramide (GalCer), which is a natural killer T (NKT) cell ligand, has been reported to exert therapeutic effects against cancer in humans and mice. Toll-like receptor (TLR) agonists systemically or locally boost antitumor efficacy in mouse cancer models. In our previous study, the co-administration of GalCer and a TLR agonist synergistically enhanced interferon-γ (IFN-γ) production in mouse splenocytes in vitro and in vivo. The increased IFN-γ production promoted a tumor antigen-specific Th1 response. Therefore, co-treatment with GalCer and a TLR agonist is expected to exert an enhanced antitumor effect. In the present study, we examined the effect of GalCer and lipopolysaccharide (LPS) combination therapy in a mouse lung-metastasis model. GalCer and LPS combination therapy markedly decreased the number of lung metastatic tumor nodes. Co-treatment with GalCer and LPS enhanced the mRNA expression of CXCL9 and CXCL10 in mediastinal lymph nodes (MLNs) and increased the number of CD8+ cells in the MLNs. Furthermore, the depletion of CD8+ T cells canceled the antitumor effect of GalCer and LPS combination therapy. Thus, GalCer and LPS combination therapy significantly enhanced tumor antigen-specific immune responses and suppressed tumor growth in a mouse lung-metastasis model.
Collapse
Affiliation(s)
- Tatsuya Ando
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Hiroyasu Ito
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Yuko Arioka
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Hideyuki Ogiso
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Mitsuru Seishima
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| |
Collapse
|
14
|
Ito H, Ando T, Ando K, Ishikawa T, Saito K, Moriwaki H, Seishima M. Induction of hepatitis B virus surface antigen-specific cytotoxic T lymphocytes can be up-regulated by the inhibition of indoleamine 2, 3-dioxygenase activity. Immunology 2014; 142:614-23. [PMID: 24580128 DOI: 10.1111/imm.12274] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/19/2014] [Accepted: 02/24/2014] [Indexed: 12/12/2022] Open
Abstract
Cytotoxic T lymphocytes (CTLs) are thought to be major effectors involved in viral clearance during acute infections, including hepatitis B virus (HBV) infection. A persistent HBV infection is characterized by a lack of or a weak CTL response to HBV, which may be reflective of tolerance to HBV. Efficient induction of HBV-specific CTLs leads to the clearance of HBV in patients with a chronic HBV infection. Previously, we reported that α-galactosylceramide (α-GalCer), a specific natural killer T (NKT) cell agonist, enhanced the induction of HBV surface antigen (HBsAg)-specific CTLs. In the present study, we found that inhibition of indoleamine 2,3-dioxygenase (IDO) activity enhanced the induction of HBsAg-specific CTLs after immunization with HBsAg and α-GalCer. The administration of HBsAg and α-GalCer increased the production of interleukin-2 and interleukin-12b, which are crucial for the induction of HBsAg-specific CTLs. The production of these cytokines was more strongly enhanced in IDO knockout mice compared with wild-type mice. In addition, α-GalCer induced the production of IDO in CD11b(+) cells, and these cells inhibited proliferation of HBsAg-specific CTLs. Our results lead to strategies for improving the induction of HBsAg-specific CTLs.
Collapse
Affiliation(s)
- Hiroyasu Ito
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Pham PV, Nguyen NT, Nguyen HM, Khuat LT, Le PM, Pham VQ, Nguyen ST, Phan NK. A simple in vitro method for evaluating dendritic cell-based vaccinations. Onco Targets Ther 2014; 7:1455-64. [PMID: 25170272 PMCID: PMC4145728 DOI: 10.2147/ott.s67057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Dendritic cell (DC) therapy is a promising therapy for cancer-targeting treatments. Recently, DCs have been used for treatment of some cancers. We aimed to develop an in vitro assay to evaluate DC therapy in cancer treatment using a breast cancer model. Methods DCs were induced from murine bone marrow mononuclear cells in Roswell Park Memorial Institute (RPMI) 1640 medium supplemented with GM-CSF (20 ng/mL) and IL-4 (20 ng/mL). Immature DCs were primed with breast cancer stem cell (BCSC)-derived antigens. BCSCs were sorted from 4T1 cell lines based on aldehyde dehydrogenase expression. A mixture of DCs and cytotoxic T lymphocytes (CTLs) were used to evaluate the inhibitory effect of antigen-primed DCs on BCSCs. BCSC proliferation and doubling time were recorded based on impedance-based cell analysis using the xCELLigence system. The specification of inhibitory effects of DCs and CTLs was also evaluated using the same system. Results The results showed that impedance-based analysis of BCSCs reflected cytotoxicity and inhibitory effects of DCs and CTLs at 72 hours. Differences in ratios of DC:CTL changed the cytotoxicity of DCs and CTLs. Conclusion This study successfully used impedance-based cell analysis as a new in vitro assay to evaluate DC efficacy in cancer immunotherapy. We hope this technique will contribute to the development and improvement of immunotherapies in the near future.
Collapse
Affiliation(s)
- Phuc Van Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Nhung Thi Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Hoang Minh Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Lam Tan Khuat
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Phong Minh Le
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Viet Quoc Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Sinh Truong Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Ngoc Kim Phan
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| |
Collapse
|
16
|
Hong Y, Peng Y, Guo ZS, Guevara-Patino J, Pang J, Butterfield LH, Mivechi N, Munn DH, Bartlett DL, He Y. Epitope-optimized alpha-fetoprotein genetic vaccines prevent carcinogen-induced murine autochthonous hepatocellular carcinoma. Hepatology 2014; 59:1448-58. [PMID: 24122861 PMCID: PMC4151349 DOI: 10.1002/hep.26893] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/25/2013] [Accepted: 10/06/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED Immunization with effective cancer vaccines can offer a much needed adjuvant therapy to fill the treatment gap after liver resection to prevent relapse of hepatocellular carcinoma (HCC). However, current HCC cancer vaccines are mostly based on native shared-self/tumor antigens that are only able to induce weak immune responses. In this study we investigated whether the HCC-associated self/tumor antigen of alpha-fetoprotein (AFP) could be engineered to create an effective vaccine to break immune tolerance and potently activate CD8 T cells to prevent clinically relevant carcinogen-induced autochthonous HCC in mice. We found that the approach of computer-guided methodical epitope-optimization created a highly immunogenic AFP and that immunization with lentivector expressing the epitope-optimized AFP, but not wild-type AFP, potently activated CD8 T cells. Critically, the activated CD8 T cells not only cross-recognized short synthetic wild-type AFP peptides, but also recognized and killed tumor cells expressing wild-type AFP protein. Immunization with lentivector expressing optimized AFP, but not native AFP, completely protected mice from tumor challenge and reduced the incidence of carcinogen-induced autochthonous HCC. In addition, prime-boost immunization with the optimized AFP significantly increased the frequency of AFP-specific memory CD8 T cells in the liver that were highly effective against emerging HCC tumor cells, further enhancing the tumor prevention of carcinogen-induced autochthonous HCC. CONCLUSIONS Epitope-optimization is required to break immune tolerance and potently activate AFP-specific CD8 T cells, generating effective antitumor effect to prevent clinically relevant carcinogen-induced autochthonous HCC in mice. Our study provides a practical roadmap to develop effective human HCC vaccines that may result in an improved outcome compared to the current HCC vaccines based on wild-type AFP.
Collapse
Affiliation(s)
- Yuan Hong
- Cancer Immunology, Inflammation, and Tolerance Program, Georgia Regents University Cancer Center, Augusta, GA
| | - Yibing Peng
- Cancer Immunology, Inflammation, and Tolerance Program, Georgia Regents University Cancer Center, Augusta, GA
| | - Z. Sheng Guo
- Department of Surgery and University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Jose Guevara-Patino
- Depart of Surgery, Cardinal Bernardin Cancer Center, Loyola University, Maywood, IL
| | - Junfeng Pang
- Department of Radiology and Molecular Chaperone Program, Georgia Regents University Cancer Center, Augusta, GA
| | - Lisa H. Butterfield
- Department of Medicine, Surgery, and Immunology, University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Nahid Mivechi
- Department of Radiology and Molecular Chaperone Program, Georgia Regents University Cancer Center, Augusta, GA
| | - David H Munn
- Cancer Immunology, Inflammation, and Tolerance Program, Georgia Regents University Cancer Center, Augusta, GA,Department of Pediatrics, Medical College of Georgia, Augusta, GA
| | - David L Bartlett
- Department of Surgery and University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Yukai He
- Cancer Immunology, Inflammation, and Tolerance Program, Georgia Regents University Cancer Center, Augusta, GA,Department of Medicine, Medical College of Georgia, Augusta, GA
| |
Collapse
|
17
|
Karimi MA, Lee E, Bachmann MH, Salicioni AM, Behrens EM, Kambayashi T, Baldwin CL. Measuring cytotoxicity by bioluminescence imaging outperforms the standard chromium-51 release assay. PLoS One 2014; 9:e89357. [PMID: 24586714 PMCID: PMC3929704 DOI: 10.1371/journal.pone.0089357] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 01/22/2014] [Indexed: 01/13/2023] Open
Abstract
The chromium-release assay developed in 1968 is still the most commonly used method to measure cytotoxicity by T cells and by natural killer cells. Target cells are loaded in vitro with radioactive chromium and lysis is determined by measuring chromium in the supernatant released by dying cells. Since then, alternative methods have been developed using different markers of target cell viability that do not involve radioactivity. Here, we compared and contrasted a bioluminescence (BLI)-based cytotoxicity assay to the standard radioactive chromium-release assay using an identical set of effector cells and tumor target cells. For this, we stably transduced several human and murine tumor cell lines to express luciferase. When co-cultured with cytotoxic effector cells, highly reproducible decreases in BLI were seen in an effector to target cell dose-dependent manner. When compared to results obtained from the chromium release assay, the performance of the BLI-based assay was superior, because of its robustness, increased signal-to-noise ratio, and faster kinetics. The reduced/delayed detection of cytotoxicity by the chromium release method was attributable to the association of chromium with structural components of the cell, which are released quickly by detergent solubilization but not by hypotonic lysis. We conclude that the (BLI)-based measurement of cytotoxicity offers a superior non-radioactive alternative to the chromium-release assay that is more robust and quicker to perform.
Collapse
Affiliation(s)
- Mobin A. Karimi
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - Eric Lee
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| | - Michael H. Bachmann
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Ana Maria Salicioni
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| | - Edward M. Behrens
- Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Cynthia L. Baldwin
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| |
Collapse
|
18
|
Watanabe T, Hotta C, Koizumi SI, Miyashita K, Nakabayashi J, Kurotaki D, Sato GR, Yamamoto M, Nakazawa M, Fujita H, Sakai R, Fujisawa S, Nishiyama A, Ikezawa Z, Aihara M, Ishigatsubo Y, Tamura T. The Transcription Factor IRF8 Counteracts BCR-ABL to Rescue Dendritic Cell Development in Chronic Myelogenous Leukemia. Cancer Res 2013; 73:6642-53. [DOI: 10.1158/0008-5472.can-13-0802] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
19
|
Harimoto H, Shimizu M, Nakagawa Y, Nakatsuka K, Wakabayashi A, Sakamoto C, Takahashi H. Inactivation of tumor-specific CD8⁺ CTLs by tumor-infiltrating tolerogenic dendritic cells. Immunol Cell Biol 2013; 91:545-55. [PMID: 24018532 PMCID: PMC3806489 DOI: 10.1038/icb.2013.38] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 07/03/2013] [Accepted: 07/07/2013] [Indexed: 02/06/2023]
Abstract
Cancer immunosurveillance failure is largely attributed to the insufficient activation of tumor-specific class I major histocompatibility complex (MHC) molecule (MHC-I)-restricted CD8+ cytotoxic T lymphocytes (CTLs). DEC-205+ dendritic cells (DCs), having the ability to cross-present, can present captured tumor antigens on MHC-I alongside costimulatory molecules, inducing the priming and activation of tumor-specific CD8+ CTLs. It has been suggested that reduced levels of costimulatory molecules on DCs may be a cause of impaired CTL induction and that some tumors may induce the downregulation of costimulatory molecules on tolerogenic DCs. To examine such possibilities, we established two distinct types of murine hepatoma cell lines, named Hepa1-6-1 and Hepa1-6-2 (derived from Hepa1-6 cells), and confirmed that they display similar antigenicities, as well as identical surface expression of MHC-I. We found that Hepa1-6-1 had the ability to grow continuously after subcutaneous implantation into syngeneic C57BL/6 mice and did not prime CD8+ CTLs. In contrast, Hepa1-6-2 cells, which display reduced levels of adhesion molecules, such as Intercellular Adhesion Molecule 1 (ICAM-1), failed to grow in vivo and efficiently primed CTLs. Moreover, Hepa1-6-1-derived factors, such as transforming growth factor (TGF)-β1, vascular endothelial growth factor (VEGF) and α-fetoprotein (AFP), converted CD11chigh MHC-IIhigh DEC-205+ DC subsets into tolerogenic cells, displaying downregulated costimulatory molecules and having impaired cross-presenting capacities. These immunosuppressive tolerogenic DCs appeared to inhibit the induction of tumor-specific CD8+ CTLs and suppress their cytotoxic functions within the tumor. Together, the findings presented here provide a new method of cancer immunotherapy using the selective suppression, depletion or alteration of immunosuppressive tolerogenic DCs within tumors.
Collapse
Affiliation(s)
- Hirotomo Harimoto
- 1] Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan [2] Third Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
20
|
Xu WL, Li SL, Wen M, Wen JY, Han J, Zhang HZ, Gao F, Cai JH. Tracking in vivo migration and distribution of antigen-specific cytotoxic T lymphocytes by 5,6-carboxyfluorescein diacetate succinimidyl ester staining during cancer immunotherapy. Chin Med J (Engl) 2013; 126:3019-3025. [PMID: 23981604 DOI: 10.3760/cma.j.issn.0366-6999.20122753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Killing of targeted tumors during adoptive cell transfer therapy is associated with cytotoxic T lymphocyte (CTL) numbers, immunophenotype, tumor-specificity, and in vivo residence time, migration, and distribution. Therefore, tracing in vivo persistence, migration, and distribution of CTLs is important for cancer immunotherapy. METHODS Optimal staining concentration for CTL proliferation was determined by cell counting kit-8 (CCK-8) assay and killing efficiencies of CTLs or carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeled melanoma antigen-specific cytotoxic T lymphocytes (CFSE-CTLs) for malignant melanoma cells in vitro were compared. Additionally, CFSE-CTLs were intravenously transfused to mice receiving B16 melanoma, and their residence time, migration, and distribution in vivo were observed by measuring fluorescence intensities of CFSE-CTLs per gram of tissue (%FI/g) in various tissues and analyzing tumor/non-tumor (T/NT) values. Anti-tumor effects of transferred CTLs and correlation between %FI/g and D-value of tumor size were analyzed. RESULTS Five-micromolar CFSE was optimal for labeling CTLs with minimal cytotoxicity. No significant difference occurred between CTLs and CFSE-CTLs for tumor cell killing (P = 0.849) or interleukin-2 (P = 0.318) and interferon-γ (P = 0.201) levels. Distribution of CTLs in vivo varied with time. A negative correlation between %FI/g in tumors and D-value of tumor sizes by Spearman correlation analysis was observed. CTLs were recruited to and killed tumors from 6 hours to 3 days after cell infusion. CTLs were observed up to three weeks later in the tumor, liver, kidneys, and spleen; this was related to the abundant blood supply or the nature of immune organs. CONCLUSIONS CCK-8 assay is a novel method to select optimal CFSE staining concentrations. Fluorescence intensity of transferred CTLs reflects their killing efficiency of tumors. CFSE fluorescent markers can trace in vivo CTL persistence, migration, and distribution because of its stability, long half-life, and low toxicity.
Collapse
Affiliation(s)
- Wei-li Xu
- Department of Pediatric Surgery, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Xu W, Cai J, Li S, Zhang H, Han J, Wen M, Wen J, Gao F. Improving the in vivo persistence, distribution and function of cytotoxic T lymphocytes by inhibiting the tumor immunosuppressive microenvironment. Scand J Immunol 2013; 78:50-60. [PMID: 23659474 DOI: 10.1111/sji.12065] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 04/12/2013] [Indexed: 11/30/2022]
Abstract
Adoptive cell transfer immunotherapy of malignant tumors has the problem of symbiosis between effector cells and tumor cells, a short in vivo residence time, and a poor killing efficiency of effector cells. Thus, releasing effector cells from the cancer immunosuppressive microenvironment and improving their effective time and functional status in vivo would seem to be ideal strategies for facilitating immunotherapy. Low-dose cyclophosphamide administration can effectively break immunotolerance by inhibiting regulatory T cells. In the present study, in order to verify whether the persistence, distribution and function of effector cells can be improved by inhibiting immunosuppressive microenvironment, low-dose cyclophosphamide was previously intraperitoneally injected into melanoma-bearing C57BL/6 mice, thereafter, CFSE-labeled cytotoxic T lymphocytes were transfused intravenously, and their effective time, distributive pattern, and killing efficiency in different groups were observed by measuring the fluorescence intensity and cell cycle of cytotoxic T lymphocytes distributed in various organs, in comparison with tumor growth. We found down-regulating Tregs in vivo can simultaneously reduce the levels of interleukin-10 and transforming growth factor-β. Migration and distribution of cytotoxic T lymphocytes in vivo was found to vary with time. Inhibition of immunotolerance can significantly improve the persistence, distribution, and function of cytotoxic T lymphocytes. Correspondingly, significantly higher secretion of perforin, granzyme B, IL-2, and IFN-γ in tumor tissues with decreased tumor growth was seen in the cyclophosphamide injection group than in the control group. Our study may provide useful information on the cyclophosphamide-mediated mechanism for facilitating tumor immunotherapy by inhibiting the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- W Xu
- Department of Pediatric Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Takahashi M, Matsumura J, Inagaki S, Takahashi H. Induction of CD56+ T cells after prolonged activation of T cells in vitro: A possible mechanism for CD4+ T-cell depletion in acquired immune deficiency syndrome patients. Hum Immunol 2011; 72:783-90. [DOI: 10.1016/j.humimm.2011.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 05/26/2011] [Accepted: 06/08/2011] [Indexed: 11/24/2022]
|