1
|
Ceuppens H, Pombo Antunes AR, Navarro L, Ertveldt T, Berdal M, Nagachinta S, De Ridder K, Lahoutte T, Keyaerts M, Devoogdt N, Goyvaerts C, D'Huyvetter M, Breckpot K. Efficient α and β - radionuclide therapy targeting fibroblast activation protein-α in an aggressive preclinical mouse tumour model. Eur J Nucl Med Mol Imaging 2025; 52:444-457. [PMID: 39237746 DOI: 10.1007/s00259-024-06914-4] [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: 06/28/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
PURPOSE Targeted radionuclide therapy (TRT) is a cancer treatment with relative therapeutic efficacy across various cancer types. We studied the therapeutic potential of TRT using fibroblast activation protein-α (FAP) targeting sdAbs (4AH29) labelled with 225Ac or 131I in immunocompetent mice in a human FAP (hFAP) expressing lung cancer mouse model. We further explored the combination of TRT with programmed cell death ligand 1 (PD-L1) immune checkpoint blockade (ICB). METHODS We studied the biodistribution and tumour uptake of [131I]I-GMIB-4AH29 and [225Ac]Ac-DOTA-4AH29 by ex vivo γ-counting. Therapeutic efficacy of [131I]I-GMIB-4AH29 and [225Ac]Ac-DOTA-4AH29 was evaluated in an immunocompetent mouse model. Flow cytometry analysis of tumours from [225Ac]Ac-DOTA-4AH29 treated mice was performed. Treatment with [225Ac]Ac-DOTA-4AH29 was repeated in combination with PD-L1 ICB. RESULTS The biodistribution showed high tumour uptake of [131I]I-GMIB-4AH29 with 3.5 ± 0.5% IA/g 1 h post-injection (p.i.) decreasing to 0.9 ± 0.1% IA/g after 24 h. Tumour uptake of [225Ac]Ac-DOTA-4AH29 was also relevant with 2.1 ± 0.5% IA/g 1 h p.i. with a less steep decrease to 1.7 ± 0.2% IA/g after 24 h. Survival was significantly improved after treatment with low and high doses [131I]I-GMIB-4AH29 or [225Ac]Ac-DOTA-4AH29 compared to vehicle solution. Moreover, we observed significantly higher PD-L1 expression in tumours of mice treated with [225Ac]Ac-DOTA-4AH29 compared to vehicle solution. Therefore, we combined high dose [225Ac]Ac-DOTA-4AH29 with PD-L1 ICB showing therapeutic synergy. CONCLUSION [225Ac]Ac-DOTA-4AH29 and [131I]I-GMIB-4AH29 exhibit high and persistent tumour targeting, translating into prolonged survival in mice bearing aggressive tumours. Moreover, we demonstrate that the combination of PD-L1 ICB with [225Ac]Ac-DOTA-4AH29 TRT enhances its therapeutic efficacy.
Collapse
Affiliation(s)
- Hannelore Ceuppens
- Vrije Universiteit Brussel, Department of Biomedical Sciences, Translational Oncology Research Center, Laboratory for Molecular and Cellular Therapy, Laarbeeklaan 103. Building E, Brussels, 1090, Belgium.
| | | | - Laurent Navarro
- Precirix NV/SA, Burg. Etienne Demunterlaan 3, Brussels, B-1090, Belgium
| | - Thomas Ertveldt
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium
| | - Marion Berdal
- Precirix NV/SA, Burg. Etienne Demunterlaan 3, Brussels, B-1090, Belgium
| | - Surasa Nagachinta
- Precirix NV/SA, Burg. Etienne Demunterlaan 3, Brussels, B-1090, Belgium
| | - Kirsten De Ridder
- Vrije Universiteit Brussel, Department of Biomedical Sciences, Translational Oncology Research Center, Laboratory for Molecular and Cellular Therapy, Laarbeeklaan 103. Building E, Brussels, 1090, Belgium
| | - Tony Lahoutte
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium
- Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel (UZ Brussel), Nuclear Medicine Department, Laarbeeklaan 101, Brussels, 1090, Belgium
| | - Marleen Keyaerts
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium
- Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel (UZ Brussel), Nuclear Medicine Department, Laarbeeklaan 101, Brussels, 1090, Belgium
| | - Nick Devoogdt
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium
| | - Cleo Goyvaerts
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium
| | - Matthias D'Huyvetter
- Precirix NV/SA, Burg. Etienne Demunterlaan 3, Brussels, B-1090, Belgium.
- Vrije Universiteit Brussel, Molecular Imaging and Therapy Research Group, Laarbeeklaan 103/K, Brussels, 1090, Belgium.
| | - Karine Breckpot
- Vrije Universiteit Brussel, Department of Biomedical Sciences, Translational Oncology Research Center, Laboratory for Molecular and Cellular Therapy, Laarbeeklaan 103. Building E, Brussels, 1090, Belgium.
| |
Collapse
|
2
|
Hanssens H, Meeus F, Gesquiere EL, Puttemans J, De Vlaeminck Y, De Veirman K, Breckpot K, Devoogdt N. Anti-Idiotypic VHHs and VHH-CAR-T Cells to Tackle Multiple Myeloma: Different Applications Call for Different Antigen-Binding Moieties. Int J Mol Sci 2024; 25:5634. [PMID: 38891821 PMCID: PMC11171536 DOI: 10.3390/ijms25115634] [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: 03/25/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
CAR-T cell therapy is at the forefront of next-generation multiple myeloma (MM) management, with two B-cell maturation antigen (BCMA)-targeted products recently approved. However, these products are incapable of breaking the infamous pattern of patient relapse. Two contributing factors are the use of BCMA as a target molecule and the artificial scFv format that is responsible for antigen recognition. Tackling both points of improvement in the present study, we used previously characterized VHHs that specifically target the idiotype of murine 5T33 MM cells. This idiotype represents one of the most promising yet challenging MM target antigens, as it is highly cancer- but also patient-specific. These VHHs were incorporated into VHH-based CAR modules, the format of which has advantages compared to scFv-based CARs. This allowed a side-by-side comparison of the influence of the targeting domain on T cell activation. Surprisingly, VHHs previously selected as lead compounds for targeted MM radiotherapy are not the best (CAR-) T cell activators. Moreover, the majority of the evaluated VHHs are incapable of inducing any T cell activation. As such, we highlight the importance of specific VHH selection, depending on its intended use, and thereby raise an important shortcoming of current common CAR development approaches.
Collapse
Affiliation(s)
- Heleen Hanssens
- Molecular Imaging and Therapy Research Group (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/K0, 1090 Brussels, Belgium; (H.H.); (E.L.G.); (J.P.)
| | - Fien Meeus
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E2, 1090 Brussels, Belgium; (F.M.); (Y.D.V.); (K.B.)
| | - Emma L. Gesquiere
- Molecular Imaging and Therapy Research Group (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/K0, 1090 Brussels, Belgium; (H.H.); (E.L.G.); (J.P.)
| | - Janik Puttemans
- Molecular Imaging and Therapy Research Group (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/K0, 1090 Brussels, Belgium; (H.H.); (E.L.G.); (J.P.)
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E2, 1090 Brussels, Belgium; (F.M.); (Y.D.V.); (K.B.)
| | - Kim De Veirman
- Laboratory for Hematology and Immunology (HEIM), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/D0, 1090 Brussels, Belgium;
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E2, 1090 Brussels, Belgium; (F.M.); (Y.D.V.); (K.B.)
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/K0, 1090 Brussels, Belgium; (H.H.); (E.L.G.); (J.P.)
| |
Collapse
|
3
|
Hanssens H, Meeus F, De Vlaeminck Y, Lecocq Q, Puttemans J, Debie P, De Groof TWM, Goyvaerts C, De Veirman K, Breckpot K, Devoogdt N. Scrutiny of chimeric antigen receptor activation by the extracellular domain: experience with single domain antibodies targeting multiple myeloma cells highlights the need for case-by-case optimization. Front Immunol 2024; 15:1389018. [PMID: 38720898 PMCID: PMC11077437 DOI: 10.3389/fimmu.2024.1389018] [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: 02/20/2024] [Accepted: 04/02/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Multiple myeloma (MM) remains incurable, despite the advent of chimeric antigen receptor (CAR)-T cell therapy. This unfulfilled potential can be attributed to two untackled issues: the lack of suitable CAR targets and formats. In relation to the former, the target should be highly expressed and reluctant to shedding; two characteristics that are attributed to the CS1-antigen. Furthermore, conventional CARs rely on scFvs for antigen recognition, yet this withholds disadvantages, mainly caused by the intrinsic instability of this format. VHHs have been proposed as valid scFv alternatives. We therefore intended to develop VHH-based CAR-T cells, targeting CS1, and to identify VHHs that induce optimal CAR-T cell activation together with the VHH parameters required to achieve this. Methods CS1-specific VHHs were generated, identified and fully characterized, in vitro and in vivo. Next, they were incorporated into second-generation CARs that only differ in their antigen-binding moiety. Reporter T-cell lines were lentivirally transduced with the different VHH-CARs and CAR-T cell activation kinetics were evaluated side-by-side. Affinity, cell-binding capacity, epitope location, in vivo behavior, binding distance, and orientation of the CAR-T:MM cell interaction pair were investigated as predictive parameters for CAR-T cell activation. Results Our data show that the VHHs affinity for its target antigen is relatively predictive for its in vivo tumor-tracing capacity, as tumor uptake generally decreased with decreasing affinity in an in vivo model of MM. This does not hold true for their CAR-T cell activation potential, as some intermediate affinity-binding VHHs proved surprisingly potent, while some higher affinity VHHs failed to induce equal levels of T-cell activation. This could not be attributed to cell-binding capacity, in vivo VHH behavior, epitope location, cell-to-cell distance or binding orientation. Hence, none of the investigated parameters proved to have significant predictive value for the extent of CAR-T cell activation. Conclusions We gained insight into the predictive parameters of VHHs in the CAR-context using a VHH library against CS1, a highly relevant MM antigen. As none of the studied VHH parameters had predictive value, defining VHHs for optimal CAR-T cell activation remains bound to serendipity. These findings highlight the importance of screening multiple candidates.
Collapse
Affiliation(s)
- Heleen Hanssens
- Laboratory of Molecular Imaging and Therapy (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
- Laboratory for Hematology and Immunology (HEIM), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fien Meeus
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Janik Puttemans
- Laboratory of Molecular Imaging and Therapy (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pieterjan Debie
- Laboratory of Molecular Imaging and Therapy (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Timo W. M. De Groof
- Laboratory of Molecular Imaging and Therapy (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Laboratory for Hematology and Immunology (HEIM), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Laboratory of Molecular Imaging and Therapy (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
4
|
Van Campenhout R, Sanz-Serrano J, Cooreman A, De Vlaeminck Y, Breckpot K, Kadam P, Tabernilla A, Vinken M. Assessment of Connexin43 Hemichannel Functionality Based on Cytosolic Uptake of Yo-Pro1. Methods Mol Biol 2024; 2801:75-85. [PMID: 38578414 DOI: 10.1007/978-1-0716-3842-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Connexin proteins are the building blocks of gap junctions and connexin hemichannels. Both provide a pathway for cellular communication. Gap junctions support intercellular communication mechanisms and regulate homeostasis. In contrast, open connexin hemichannels connect the intracellular compartment and the extracellular environment, and their activation fuels inflammation and cell death. The development of clinically applicable connexin hemichannel blockers for therapeutic purposes is therefore gaining momentum. This chapter describes a well-established protocol optimized for assessing connexin hemichannel activity by using the reporter dye Yo-Pro1.
Collapse
Affiliation(s)
- Raf Van Campenhout
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Julen Sanz-Serrano
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Axelle Cooreman
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Prashant Kadam
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Andrés Tabernilla
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mathieu Vinken
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
5
|
Awad RM, De Vlaeminck Y, Meeus F, Ertveldt T, Zeven K, Ceuppens H, Goyvaerts C, Verdonck M, Salguero G, Raes G, Devoogdt N, Breckpot K. In vitro modelling of local gene therapy with IL-15/IL-15Rα and a PD-L1 antagonist in melanoma reveals an interplay between NK cells and CD4 + T cells. Sci Rep 2023; 13:18995. [PMID: 37923822 PMCID: PMC10624833 DOI: 10.1038/s41598-023-45948-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023] Open
Abstract
Blockade of the immune checkpoint axis consisting of programmed death-1 (PD-1) and its ligand PD-L1 alleviates the functional inhibition of tumor-infiltrating lymphoid cells yet weakly induces their expansion. Exogenous cytokines could further expand lymphoid cells and thus synergize with αPD-L1 therapy. However, systemic delivery of most cytokines causes severe toxicity due to unspecific expansion of immune cells in the periphery. Here, we modelled local delivery of cytokines and αPD-L1 therapeutics to immune cell-containing in vitro melanoma tumors. Three-dimensional tumor models consisting of 624-MEL cells were co-cultured with human peripheral blood lymphoid cells (PBLs) in presence of the cytokines IL-2, IL-7, IL-15, IL-21 and IFN-γ. To model local gene therapy, melanoma tumors were modified with lentiviral vectors encoding IL-15 fused to IL-15Rα (IL-15/IL-15Rα) and K2-Fc, a fusion of a human PD-L1 specific single domain antibody to immunoglobulin (Ig)G1 Fc. To evaluate the interplay between PBL fractions, NK cells, CD4+ T cells or CD8+ T cells were depleted. Tumor cell killing was followed up using real time imaging and immune cell expansion and activation was evaluated with flow cytometry. Among the tested cytokines, IL-15 was the most potent cytokine in stimulating tumor cell killing and expanding both natural killer (NK) cells and CD8+ T cells. Gene-based delivery of IL-15/IL-15Rα to tumor cells, shows expansion of NK cells, activation of NK cells, CD4+ and CD8+ T cells, and killing of tumor spheroids. Both NK cells and CD8+ T cells are necessary for tumor cell killing and CD4+ T-cell activation was reduced without NK cells. Co-delivery of K2-Fc improved tumor cell killing coinciding with increased activation of NK cells, which was independent of bystander T cells. CD4+ or CD8+ T cells were not affected by the co-delivery of K2-Fc even though NK-cell activation impacted CD4+ T-cell activation. This study demonstrates that gene-based delivery of IL-15/IL-15Rα to tumor cells effectively mediates anti-tumor activity and sensitizes the tumor microenvironment for therapy with αPD-L1 therapeutics mainly by impacting NK cells. These findings warrant further investigation of gene-based IL-15 and K2-Fc delivery in vivo.
Collapse
Affiliation(s)
- Robin Maximilian Awad
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium.
| | - Yannick De Vlaeminck
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Fien Meeus
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Thomas Ertveldt
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Katty Zeven
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Hannelore Ceuppens
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Cleo Goyvaerts
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Magali Verdonck
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Gustavo Salguero
- Advanced Therapies Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud-IDCBIS, 111611, Bogotá, Colombia
| | - Geert Raes
- Laboratory of Cellular and Molecular Immunology, Department of Bioengineering Sciences, Vrije Universiteit Brussel, 1050, Brussels, Belgium
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050, Brussels, Belgium
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, 1050, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Karine Breckpot
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences (BMWE), Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium.
| |
Collapse
|
6
|
Zeven K, De Groof TW, Ceuppens H, Awad RM, Ertveldt T, de Mey W, Meeus F, Raes G, Breckpot K, Devoogdt N. Development and evaluation of nanobody tracers for noninvasive nuclear imaging of the immune-checkpoint TIGIT. Front Immunol 2023; 14:1268900. [PMID: 37799715 PMCID: PMC10548220 DOI: 10.3389/fimmu.2023.1268900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/04/2023] [Indexed: 10/07/2023] Open
Abstract
Introduction T cell Ig and ITIM domain receptor (TIGIT) is a next-generation immune checkpoint predominantly expressed on activated T cells and NK cells, exhibiting an unfavorable prognostic association with various malignancies. Despite the emergence of multiple TIGIT-blocking agents entering clinical trials, only a fraction of patients responded positively to anti-TIGIT therapy. Consequently, an urgent demand arises for noninvasive techniques to quantify and monitor TIGIT expression, facilitating patient stratification and enhancing therapeutic outcomes. Small antigen binding moieties such as nanobodies, are promising candidates for such tracer development. Methods We generated a panel of anti-human or anti-mouse TIGIT nanobodies from immunized llamas. In addition, we designed a single-chain variable fragment derived from the clinically tested monoclonal antibody Vibostolimab targeting TIGIT, and assessed its performance alongside the nanobodies. In vitro characterization studies were performed, including binding ability and affinity to cell expressed or recombinant TIGIT. After Technetium-99m labeling, the nanobodies and the single-chain variable fragment were evaluated in vivo for their ability to detect TIGIT expression using SPECT/CT imaging, followed by ex vivo biodistribution analysis. Results Nine nanobodies were selected for binding to recombinant and cell expressed TIGIT with low sub-nanomolar affinities and are thermostable. A six-fold higher uptake in TIGIT-overexpressing tumor was demonstrated one hour post- injection with Technetium-99m labeled nanobodies compared to an irrelevant control nanobody. Though the single-chain variable fragment exhibited superior binding to TIGIT-expressing peripheral blood mononuclear cells in vitro, its in vivo behavior yielded lower tumor-to-background ratios at one hour post- injection, indicating that nanobodies are better suited for in vivo imaging than the single-chain variable fragment. Despite the good affinity, high specificity and on-target uptake in mice in this setting, imaging of TIGIT expression on tumor- infiltrating lymphocytes within MC38 tumors remained elusive. This is likely due to the low expression levels of TIGIT in this model. Discussion The excellent affinity, high specificity and rapid on-target uptake in mice bearing TIGIT- overexpressing tumors showed the promising diagnostic potential of nanobodies to noninvasively image high TIGIT expression within the tumor. These findings hold promise for clinical translation to aid patient selection and improve therapy response.
Collapse
Affiliation(s)
- Katty Zeven
- Laboratory of Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Timo W.M. De Groof
- Laboratory of Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Hannelore Ceuppens
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Wout de Mey
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Fien Meeus
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Geert Raes
- Laboratory for Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Myeloid Cell Immunology Lab, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Nick Devoogdt
- Laboratory of Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
7
|
De Beck L, Awad RM, Basso V, Casares N, De Ridder K, De Vlaeminck Y, Gnata A, Goyvaerts C, Lecocq Q, San José-Enériz E, Verhulst S, Maes K, Vanderkerken K, Agirre X, Prosper F, Lasarte JJ, Mondino A, Breckpot K. Inhibiting Histone and DNA Methylation Improves Cancer Vaccination in an Experimental Model of Melanoma. Front Immunol 2022; 13:799636. [PMID: 35634329 PMCID: PMC9134079 DOI: 10.3389/fimmu.2022.799636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy has improved the treatment of malignant skin cancer of the melanoma type, yet overall clinical response rates remain low. Combination therapies could be key to meet this cogent medical need. Because epigenetic hallmarks represent promising combination therapy targets, we studied the immunogenic potential of a dual inhibitor of histone methyltransferase G9a and DNA methyltransferases (DNMTs) in the preclinical B16-OVA melanoma model. Making use of tumor transcriptomic and functional analyses, methylation-targeted epigenetic reprogramming was shown to induce tumor cell cycle arrest and apoptosis in vitro coinciding with transient tumor growth delay and an IFN-I response in immune-competent mice. In consideration of a potential impact on immune cells, the drug was shown not to interfere with dendritic cell maturation or T-cell activation in vitro. Notably, the drug promoted dendritic cell and, to a lesser extent, T-cell infiltration in vivo, yet failed to sensitize tumor cells to programmed cell death-1 inhibition. Instead, it increased therapeutic efficacy of TCR-redirected T cell and dendritic cell vaccination, jointly increasing overall survival of B16-OVA tumor-bearing mice. The reported data confirm the prospect of methylation-targeted epigenetic reprogramming in melanoma and sustain dual G9a and DNMT inhibition as a strategy to tip the cancer-immune set-point towards responsiveness to active and adoptive vaccination against melanoma.
Collapse
Affiliation(s)
- Lien De Beck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Veronica Basso
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Noelia Casares
- Immunology and Immunotherapy Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, Pamplona, Spain
| | - Kirsten De Ridder
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Alessandra Gnata
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Edurne San José-Enériz
- Hemato-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, Pamplona, Spain
| | - Stefaan Verhulst
- Liver Cell Biology Research Group, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Ken Maes
- Laboratory of Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Center for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Karin Vanderkerken
- Laboratory of Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Xabier Agirre
- Hemato-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, Pamplona, Spain
- Laboratory of Cancer Epigenetics, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Felipe Prosper
- Hemato-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, Pamplona, Spain
- Laboratory of Cancer Epigenetics, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Juan José Lasarte
- Immunology and Immunotherapy Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, Pamplona, Spain
| | - Anna Mondino
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
8
|
Lecocq Q, Awad RM, De Vlaeminck Y, de Mey W, Ertveldt T, Goyvaerts C, Raes G, Thielemans K, Keyaerts M, Devoogdt N, Breckpot K. Single-Domain Antibody Nuclear Imaging Allows Noninvasive Quantification of LAG-3 Expression by Tumor-Infiltrating Leukocytes and Predicts Response of Immune Checkpoint Blockade. J Nucl Med 2021; 62:1638-1644. [PMID: 33712537 PMCID: PMC8612328 DOI: 10.2967/jnumed.120.258871] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/03/2021] [Indexed: 11/29/2022] Open
Abstract
Recent advances in the field of immune-oncology led to the discovery of next-generation immune checkpoints (ICPs). Lymphocyte activation gene-3 (LAG-3), being the most widely studied among them, is being explored as a target for the treatment of cancer patients. Several antagonistic anti-LAG-3 antibodies are being developed and are prime candidates for clinical application. Furthermore, validated therapies targeting cytotoxic T-lymphocyte-associated protein-4, programmed cell-death protein-1, or programmed cell-death ligand-1 showed that only subsets of patients respond. This finding highlights the need for better tools for patient selection and monitoring. The potential of molecular imaging to detect ICPs noninvasively in cancer is supported by several preclinical and clinical studies. Here, we report on a single-domain antibody to evaluate whole-body LAG-3 expression in various syngeneic mouse cancer models using nuclear imaging. Methods: SPECT/CT scans of tumor-bearing mice were performed 1 h after injection with radiolabeled single-domain antibody. Organs and tumors of mice were isolated and evaluated for the presence of the radiolabeled tracer and LAG-3-expressing immune cells using a γ-counter and flow cytometry respectively. PD-1/LAG-3-blocking antibodies were injected in MC38-bearing mice. Results: The radiolabeled single-domain antibody detected LAG-3 expression on tumor-infiltrating lymphocytes (TILs) as soon as 1 h after injection in MC38, MO4, and TC-1 cancer models. The single-domain antibody tracer visualized a compensatory upregulation of LAG-3 on TILs in MC38 tumors of mice treated with PD-1-blocking antibodies. When PD-1 blockade was combined with LAG-3 blockade, a synergistic effect on tumor growth delay was observed. Conclusion: These findings consolidate LAG-3 as a next-generation ICP and support the use of single-domain antibodies as tools to noninvasively monitor the dynamic evolution of LAG-3 expression by TILs, which could be exploited to predict therapy outcome.
Collapse
Affiliation(s)
- Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wout de Mey
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Raes
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
- Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kris Thielemans
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium; and
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
9
|
Awad RM, Lecocq Q, Zeven K, Ertveldt T, De Beck L, Ceuppens H, Broos K, De Vlaeminck Y, Goyvaerts C, Verdonck M, Raes G, Van Parys A, Cauwels A, Keyaerts M, Devoogdt N, Breckpot K. Formatting and gene-based delivery of a human PD-L1 single domain antibody for immune checkpoint blockade. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 22:172-182. [PMID: 34485603 PMCID: PMC8397838 DOI: 10.1016/j.omtm.2021.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 05/27/2021] [Indexed: 12/03/2022]
Abstract
Monoclonal antibodies that target the inhibitory immune checkpoint axis consisting of programmed cell death protein 1 (PD-1) and its ligand, PD-L1, have changed the immune-oncology field. We identified K2, an anti-human PD-L1 single-domain antibody fragment, that can enhance T cell activation and tumor cell killing. In this study, the potential of different K2 formats as immune checkpoint blocking medicines was evaluated using a gene-based delivery approach. We showed that 2K2 and 3K2, a bivalent and trivalent K2 format generated using a 12 GS (glycine-serine) linker, were 313- and 135-fold more potent in enhancing T cell receptor (TCR) signaling in PD-1POS cells than was monovalent K2. We further showed that bivalent constructs generated using a 30 GS linker or disulfide bond were 169- and 35-fold less potent in enhancing TCR signaling than was 2K2. 2K2 enhanced tumor cell killing in a 3D melanoma model, albeit to a lesser extent than avelumab. Therefore, an immunoglobulin (Ig)G1 antibody-like fusion protein was generated, referred to as K2-Fc. K2-Fc was significantly better than avelumab in enhancing tumor cell killing in the 3D melanoma model. Overall, this study describes K2-based immune checkpoint medicines, and it highlights the benefit of an IgG1 Fc fusion to K2 that gains bivalency, effector functions, and efficacy.
Collapse
Affiliation(s)
- Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Katty Zeven
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium.,In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Lien De Beck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Hannelore Ceuppens
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Katrijn Broos
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Magali Verdonck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Geert Raes
- Laboratory of Cellular and Molecular Immunology, Department of Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium.,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, 1050 Brussels, Belgium
| | - Alexander Van Parys
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Anje Cauwels
- Cytokine Receptor Laboratory, Flanders Institute of Biotechnology, VIB Medical Biotechnology Center, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Marleen Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, 1090 Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, 1090 Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| |
Collapse
|
10
|
De Vlaeminck Y, Bonelli S, Awad RM, Dewilde M, Rizzolio S, Lecocq Q, Bolli E, Santos AR, Laoui D, Schoonooghe S, Tamagnone L, Goyvaerts C, Mazzone M, Breckpot K, Van Ginderachter JA. Targeting Neuropilin-1 with Nanobodies Reduces Colorectal Carcinoma Development. Cancers (Basel) 2020; 12:cancers12123582. [PMID: 33266104 PMCID: PMC7760077 DOI: 10.3390/cancers12123582] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Neuropilin-1 is a co-receptor for semaphorins and vascular endothelial growth factor family members. Neuropilin-1 can be expressed on tumor cells, tumor-infiltrating myeloid and lymphoid cells and has been linked to a tumor-promoting environment. We investigated nanobodies (Nbs) targeting neuropilin-1 for their potential to hamper colorectal carcinoma development in mice. Our data suggest that targeting neuropilin-1 in cancer using neuropilin-1 blocking Nbs delays tumor growth and extends the survival through a shift in the anti-tumor macrophage/pro-tumor macrophage ratio and activation of colorectal cancer-specific CD8+ T cells. These findings provide a rationale for the further development of Nbs targeting human neuropilin-1 and bringing them from the bench to the bedside. Abstract Neuropilin-1 (NRP-1) is a co-receptor for semaphorins and vascular endothelial growth factor (VEGF) family members that can be expressed on cancer cells and tumor-infiltrating myeloid, endothelial and lymphoid cells. It has been linked to a tumor-promoting environment upon interaction with semaphorin 3A (Sema3A). Nanobodies (Nbs) targeting NRP-1 were generated for their potential to hamper the NRP-1/Sema3A interaction and their impact on colorectal carcinoma (CRC) development was evaluated in vivo through the generation of anti-NRP-1-producing CRC cells. We observed that tumor growth was significantly delayed and survival prolonged when the anti-NRP-1 Nbs were produced in vivo. We further analyzed the tumor microenvironment and observed that the pro-inflammatory MHC-IIhigh/trophic MHC-IIlow macrophage ratio was increased in tumors that produce anti-NRP-1 Nbs. This finding was corroborated by an increase in the expression of genes associated with MHC-IIhigh macrophages and a decrease in the expression of MHC-IIlow macrophage-associated genes in the macrophage pool sorted from anti-NRP-1 Nb-producing tumors. Moreover, we observed a significantly higher percentage of tumor-associated antigen-specific CD8+ T cells in tumors producing anti-NRP-1 Nbs. These data demonstrate that an intratumoral expression of NRP-1/Sema3A blocking biologicals increases anti-tumor immunity.
Collapse
Affiliation(s)
- Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (Y.D.V.); (R.M.A.); (Q.L.); (C.G.)
| | - Stefano Bonelli
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1040 Brussels, Belgium; (S.B.); (E.B.); (D.L.); (S.S.)
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, 1040 Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (Y.D.V.); (R.M.A.); (Q.L.); (C.G.)
| | - Maarten Dewilde
- VIB Discovery Sciences, 3000 Leuven, Belgium; (M.D.); (A.R.S.)
| | | | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (Y.D.V.); (R.M.A.); (Q.L.); (C.G.)
| | - Evangelia Bolli
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1040 Brussels, Belgium; (S.B.); (E.B.); (D.L.); (S.S.)
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, 1040 Brussels, Belgium
| | - Ana Rita Santos
- VIB Discovery Sciences, 3000 Leuven, Belgium; (M.D.); (A.R.S.)
| | - Damya Laoui
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1040 Brussels, Belgium; (S.B.); (E.B.); (D.L.); (S.S.)
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, 1040 Brussels, Belgium
| | - Steve Schoonooghe
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1040 Brussels, Belgium; (S.B.); (E.B.); (D.L.); (S.S.)
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, 1040 Brussels, Belgium
| | - Luca Tamagnone
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00100 Rome, Italy;
- Department of Oncology, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00100 Rome, Italy
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (Y.D.V.); (R.M.A.); (Q.L.); (C.G.)
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, VIB Center for Cancer Biology, 3000 Leuven, Belgium;
- Department of Oncology, Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, KU Leuven, 3000 Leuven, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (Y.D.V.); (R.M.A.); (Q.L.); (C.G.)
- Correspondence: (K.B.); (J.A.V.G.)
| | - Jo A. Van Ginderachter
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1040 Brussels, Belgium; (S.B.); (E.B.); (D.L.); (S.S.)
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, 1040 Brussels, Belgium
- Correspondence: (K.B.); (J.A.V.G.)
| |
Collapse
|
11
|
Harizaj A, De Smedt SC, Lentacker I, Braeckmans K. Physical transfection technologies for macrophages and dendritic cells in immunotherapy. Expert Opin Drug Deliv 2020; 18:229-247. [PMID: 32985919 DOI: 10.1080/17425247.2021.1828340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Dendritic cells (DCs) and macrophages, two important antigen presenting cells (APCs) of the innate immune system, are being explored for the use in cell-based cancer immunotherapy. For this application, the therapeutic potential of patient-derived APCs is increased by delivering different types of functional macromolecules, such as mRNA and pDNA, into their cytosol. Compared to the use of viral and non-viral delivery vectors, physical intracellular delivery techniques are known to be more straightforward, more controllable, faster and generate high delivery efficiencies. AREAS COVERED This review starts with electroporation as the most traditional physical transfection method, before continuing with the more recent technologies such as sonoporation, nanowires and microfluidic cell squeezing. A description is provided of each of those intracellular delivery technologies with their strengths and weaknesses, especially paying attention to delivery efficiency and safety profile. EXPERT OPINION Given the common use of electroporation for the production of therapeutic APCs, it is recommended that more detailed studies are performed on the effect of electroporation on APC fitness, even down to the genetic level. Newer intracellular delivery technologies seem to have less impact on APC functionality but further work is needed to fully uncover their suitability to transfect APCs with different types of macromolecules.
Collapse
Affiliation(s)
- Aranit Harizaj
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Ine Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| |
Collapse
|
12
|
Hypoxia-Induced Glioma-Derived Exosomal miRNA-199a-3p Promotes Ischemic Injury of Peritumoral Neurons by Inhibiting the mTOR Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5609637. [PMID: 33110474 PMCID: PMC7578720 DOI: 10.1155/2020/5609637] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/04/2020] [Indexed: 11/18/2022]
Abstract
The underlying molecular mechanisms that the hypoxic microenvironment could aggravate neuronal injury are still not clear. In this study, we hypothesized that the exosomes, exosomal miRNAs, and the mTOR signaling pathway might be involved in hypoxic peritumoral neuronal injury in glioma. Multimodal radiological images, HE, and HIF-1α staining of high-grade glioma (HGG) samples revealed that the peritumoral hypoxic area overlapped with the cytotoxic edema region and directly contacted with normal neurons. In either direct or indirect coculture system, hypoxia could promote normal mouse hippocampal neuronal cell (HT22) injury, and the growth of HT22 cells was suppressed by C6 glioma cells under hypoxic condition. For administrating hypoxia-induced glioma-derived exosomes (HIGDE) that could aggravate oxygen-glucose deprivation (OGD)/reperfusion neuronal injury, we identified that exosomes may be the communication medium between glioma cells and peritumoral neurons, and we furtherly found that exosomal miR-199a-3p mediated the OGD/reperfusion neuronal injury process by suppressing the mTOR signaling pathway. Moreover, the upregulation of miRNA-199a-3p in exosomes from glioma cells was induced by hypoxia-related HIF-1α activation. To sum up, hypoxia-induced glioma-derived exosomal miRNA-199a-3p can be upregulated by the activation of HIF-1α and is able to increase the ischemic injury of peritumoral neurons by inhibiting the mTOR pathway.
Collapse
|
13
|
Das B, Senapati S. Immunological and functional aspects of MAGEA3 cancer/testis antigen. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 125:121-147. [PMID: 33931137 DOI: 10.1016/bs.apcsb.2020.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Identification of ectopic gene activation in cancer cells serves as a basis for both gene signature-guided tumor targeting and unearthing of oncogenic mechanisms to expand the understanding of tumor biology/oncogenic process. Proteins expressed only in germ cells of testis and/or placenta (immunoprivileged organs) and in malignancies are called cancer testis antigens; they are antigenic because of the lack of antigen presentation by those specific cell types (germ cells), which limits the exposure of the proteins to the immune cells. Since the Cancer Testis Antigens (CTAs) are immunogenic and expressed in a wide variety of cancer types, CT antigens have become interesting target for immunotherapy against cancer. Among CT antigens MAGEA family is reported to have 12 members (MAGEA1 to MAGEA12). The current review highlights the studies on MAGEA3 which is a CT antigen and reported in almost all types of cancer. MAGEA3 is well tried for cancer immunotherapy. Recent advances on its functional and immunological aspect warranted much deliberation on effective therapeutic approach, thus making it a more interesting target for cancer therapy.
Collapse
Affiliation(s)
- Biswajit Das
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India; Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shantibhusan Senapati
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India.
| |
Collapse
|
14
|
Madley R, Nauman G, Danzl N, Borsotti C, Khosravi Maharlooei M, Li HW, Chavez E, Creusot RJ, Nakayama M, Roep B, Sykes M. Negative selection of human T cells recognizing a naturally-expressed tissue-restricted antigen in the human thymus. J Transl Autoimmun 2020; 3:100061. [PMID: 32875283 PMCID: PMC7451786 DOI: 10.1016/j.jtauto.2020.100061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/02/2020] [Indexed: 12/15/2022] Open
Abstract
During T cell development in mice, thymic negative selection deletes cells with the potential to recognize and react to self-antigens. In human T cell-dependent autoimmune diseases such as Type 1 diabetes, multiple sclerosis, and rheumatoid arthritis, T cells reactive to autoantigens are thought to escape negative selection, traffic to the periphery and attack self-tissues. However, physiological thymic negative selection of autoreactive human T cells has not been previously studied. We now describe a human T-cell receptor-transgenic humanized mouse model that permits the study of autoreactive T-cell development in a human thymus. Our studies demonstrate that thymocytes expressing the autoreactive Clone 5 TCR, which recognizes insulin B:9-23 presented by HLA-DQ8, are efficiently negatively selected at the double and single positive stage in human immune systems derived from HLA-DQ8+ HSCs. In the absence of hematopoietic expression of the HLA restriction element, negative selection of Clone 5 is less efficient and restricted to the single positive stage. To our knowledge, these data provide the first demonstration of negative selection of human T cells recognizing a naturally-expressed tissue-restricted antigen. Intrathymic antigen presenting cells are required to delete less mature thymocytes, while presentation by medullary thymic epithelial cells may be sufficient to delete more mature single positive cells. These observations set the stage for investigation of putative defects in negative selection in human autoimmune diseases.
Collapse
Affiliation(s)
- Rachel Madley
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA,Columbia University Department of Microbiology and Immunology, New York, NY, 10032, USA
| | - Grace Nauman
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA,Columbia University Department of Microbiology and Immunology, New York, NY, 10032, USA
| | - Nichole Danzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Chiara Borsotti
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Mohsen Khosravi Maharlooei
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Hao Wei Li
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Estefania Chavez
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Remi J. Creusot
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Bart Roep
- Department of Immunohaematology & Blood Transfusion, Leiden University Medical Center, 2300 RC, Leiden, the Netherlands,Department of Diabetes Immunology, Diabetes & Metabolism Research Institute at the Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA,Columbia University Department of Microbiology and Immunology, New York, NY, 10032, USA,Columbia University Department of Surgery, New York, NY, 10032, USA,Corresponding author. Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA.
| |
Collapse
|
15
|
Norton TD, Tada T, Leibowitz R, van der Heide V, Homann D, Landau NR. Lentiviral-Vector-Based Dendritic Cell Vaccine Synergizes with Checkpoint Blockade to Clear Chronic Viral Infection. Mol Ther 2020; 28:1795-1805. [PMID: 32497512 DOI: 10.1016/j.ymthe.2020.05.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/13/2020] [Accepted: 05/14/2020] [Indexed: 12/17/2022] Open
Abstract
Dendritic cell vaccines are a promising strategy for the treatment of cancer and infectious diseases but have met with mixed success. We report on a lentiviral vector-based dendritic cell vaccine strategy that generates a cluster of differentiation 8 (CD8) T cell response that is much stronger than that achieved by standard peptide-pulsing approaches. The strategy was tested in the mouse lymphocytic choriomeningitis virus (LCMV) model. Bone marrow-derived dendritic cells from SAMHD1 knockout mice were transduced with a lentiviral vector expressing the GP33 major-histocompatibility-complex (MHC)-class-I-restricted peptide epitope and CD40 ligand (CD40L) and injected into wild-type mice. The mice were highly protected against acute and chronic variant CL-13 LCMVs, resulting in a 100-fold greater decrease than that achieved with peptide epitope-pulsed dendritic cells. Inclusion of an MHC-class-II-restricted epitope in the lentiviral vector further increased the CD8 T cell response and resulted in antigen-specific CD8 T cells that exhibited a phenotype associated with functional cytotoxic T cells. The vaccination synergized with checkpoint blockade to reduce the viral load of mice chronically infected with CL-13 to an undetectable level. The strategy improves upon current dendritic cell vaccine strategies; is applicable to the treatment of disease, including AIDS and cancer; and supports the utility of Vpx-containing vectors.
Collapse
Affiliation(s)
- Thomas D Norton
- Department of Medicine, Division of Infectious Diseases, New York University Langone Medical Center, New York, NY 10016, USA; Department of Microbiology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Takuya Tada
- Department of Medicine, Division of Infectious Diseases, New York University Langone Medical Center, New York, NY 10016, USA
| | - Rebecca Leibowitz
- Department of Microbiology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Verena van der Heide
- Diabetes, Obesity and Metabolism Institute & Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dirk Homann
- Diabetes, Obesity and Metabolism Institute & Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nathaniel R Landau
- Department of Microbiology, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
16
|
Preclinical Targeted α- and β --Radionuclide Therapy in HER2-Positive Brain Metastasis Using Camelid Single-Domain Antibodies. Cancers (Basel) 2020; 12:cancers12041017. [PMID: 32326199 PMCID: PMC7226418 DOI: 10.3390/cancers12041017] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022] Open
Abstract
HER2-targeted therapies have drastically improved the outcome for breast cancer patients. However, when metastasis to the brain is involved, current strategies fail to hold up to the same promise. Camelid single-domain antibody-fragments (sdAbs) have been demonstrated to possess favorable properties for detecting and treating cancerous lesions in vivo using different radiolabeling methods. Here we evaluate the anti-HER2 sdAb 2Rs15d, coupled to diagnostic γ- and therapeutic α- and β−-emitting radionuclides for the detection and treatment of HER2pos brain lesions in a preclinical setting. 2Rs15d was radiolabeled with 111In, 225Ac and 131I using DTPA- and DOTA-based bifunctional chelators and Sn-precursor of SGMIB respectively and evaluated in orthotopic tumor-bearing athymic nude mice. Therapeutic efficacy as well as systemic toxicity were determined for 131I- and 225Ac-labeled sdAbs and compared to anti-HER2 monoclonal antibody (mAb) trastuzumab in two different HER2pos tumor models. Radiolabeled 2Rs15d showed high and specific tumor uptake in both HER2pos SK-OV-3-Luc-IP1 and HER2pos MDA-MB-231Br brain lesions, whereas radiolabeled trastuzumab was unable to accumulate in intracranial SK-OV-3-Luc-IP1 tumors. Administration of [131I]-2Rs15d and [225Ac]-2Rs15d alone and in combination with trastuzumab showed a significant increase in median survival in 2 tumor models that remained largely unresponsive to trastuzumab treatment alone. Histopathological analysis revealed no significant early toxicity. Radiolabeled sdAbs prove to be promising vehicles for molecular imaging and targeted radionuclide therapy of metastatic lesions in the brain. These data demonstrate the potential of radiolabeled sdAbs as a valuable add-on treatment option for patients with difficult-to-treat HER2pos metastatic cancer.
Collapse
|
17
|
Nauman G, Borsotti C, Danzl N, Khosravi-Maharlooei M, Li HW, Chavez E, Stone S, Sykes M. Reduced positive selection of a human TCR in a swine thymus using a humanized mouse model for xenotolerance induction. Xenotransplantation 2020; 27:e12558. [PMID: 31565822 PMCID: PMC7007369 DOI: 10.1111/xen.12558] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/27/2019] [Accepted: 09/13/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Tolerance-inducing approaches to xenotransplantation would be optimal and may be necessary for long-term survival of transplanted pig organs in human patients. The ideal approach would generate donor-specific unresponsiveness to the pig organ without suppressing the patient's normal immune function. Porcine thymus transplantation has shown efficacy in promoting xenotolerance in humanized mice and large animal models. However, murine studies demonstrate that T cells selected in a swine thymus are positively selected only by swine thymic epithelial cells, and therefore, cells expressing human HLA-restricted TCRs may not be selected efficiently in a transplanted pig thymus. This may lead to suboptimal patient immune function. METHODS To assess human thymocyte selection in a pig thymus, we used a TCR transgenic humanized mouse model to study positive selection of cells expressing the MART1 TCR, a well-characterized human HLA-A2-restricted TCR, in a grafted pig thymus. RESULTS Positive selection of T cells expressing the MART1 TCR was inefficient in both a non-selecting human HLA-A2- or swine thymus compared with an HLA-A2+ thymus. Additionally, CD8 MART1 TCRbright T cells were detected in the spleens of mice transplanted with HLA-A2+ thymi but were significantly reduced in the spleens of mice transplanted with swine or HLA-A2- thymi. [Correction added on October 15, 2019, after first online publication: The missing superscript values +, -, and bright have been included in the Results section.] CONCLUSIONS: Positive selection of cells expressing a human-restricted TCR in a transplanted pig thymus is inefficient, suggesting that modifications to improve positive selection of cells expressing human-restricted TCRs in a pig thymus may be necessary to support development of a protective human T-cell pool in future patients.
Collapse
Affiliation(s)
- Grace Nauman
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Chiara Borsotti
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Nichole Danzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Mohsen Khosravi-Maharlooei
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Hao-Wei Li
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Estefania Chavez
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Samantha Stone
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia University, New York, NY, USA
- Department of Surgery, Columbia University Medical Center, Columbia University, New York, NY, USA
| |
Collapse
|
18
|
Noninvasive Imaging of the Immune Checkpoint LAG-3 Using Nanobodies, from Development to Pre-Clinical Use. Biomolecules 2019; 9:biom9100548. [PMID: 31569553 PMCID: PMC6843898 DOI: 10.3390/biom9100548] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoint inhibition (ICI) is a promising cancer therapy, which has progressed rapidly from a preclinical concept to clinical implementation. Commonly considered targets in ICI are CTLA-4, PD-1/PD-L1, and LAG-3, and the list grows. As ICI is generally only beneficial for a subset of patients, there is a need to select patients that are eligible for therapy as well as to monitor therapy response. There is growing interest to do this noninvasively, by molecular imaging with target-specific tracers. To this day, noninvasive imaging has focused on CTLA-4 and PD-1/PD-L1, while there is no noninvasive tool available to accurately assess LAG-3 expression in vivo. In this proof-of-concept study, we developed nanobodies, the smallest functional fragments from camelid heavy chain-only antibodies, to noninvasively evaluate mouse LAG-3 expression using single photon emission computed tomography (SPECT)/CT imaging. The in vitro characterization of 114 nanobodies led to the selection of nine nanobodies binding to mouse LAG-3. The injection of 99mTechnetium-labeled nanobodies in healthy mice showed specific uptake in immune peripheral organs like the spleen and lymph nodes, which was not observed in LAG-3 gene knock-out mice. Moreover, nanobody uptake could be visualized using SPECT/CT and correlated to the presence of LAG-3 as assessed in flow cytometry and immunohistochemistry. SPECT/CT scans of tumor bearing mice further confirmed the diagnostic potential of the nanobodies. These findings substantiate the approach to use nanobodies as a tool to image inhibitory immune checkpoints in the tumor environment.
Collapse
|
19
|
Single Domain Antibody-Mediated Blockade of Programmed Death-Ligand 1 on Dendritic Cells Enhances CD8 T-cell Activation and Cytokine Production. Vaccines (Basel) 2019; 7:vaccines7030085. [PMID: 31394834 PMCID: PMC6789804 DOI: 10.3390/vaccines7030085] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/01/2019] [Accepted: 08/03/2019] [Indexed: 12/22/2022] Open
Abstract
Dendritic cell [DC] vaccines can induce durable clinical responses, at least in a fraction of previously treated, late stage cancer patients. Several preclinical studies suggest that shielding programmed death-ligand 1 [PD-L1] on the DC surface may be an attractive strategy to extend such clinical benefits to a larger patient population. In this study, we evaluated the use of single domain antibody [sdAb] K2, a high affinity, antagonistic, PD-L1 specific sdAb, for its ability to enhance DC mediated T-cell activation and benchmarked it against the use of the monoclonal antibodies [mAbs], MIH1, 29E.2A3 and avelumab. Similar to mAbs, sdAb K2 enhanced antigen-specific T-cell receptor signaling in PD-1 positive (PD-1pos) reporter cells activated by DCs. We further showed that the activation and function of antigen-specific CD8 positive (CD8pos) T cells, activated by DCs, was enhanced by inclusion of sdAb K2, but not mAbs. The failure of mAbs to enhance T-cell activation might be explained by their low efficacy to bind PD-L1 on DCs when compared to binding of PD-L1 on non-immune cells, whereas sdAb K2 shows high binding to PD-L1 on immune as well as non-immune cells. These data provide a rationale for the inclusion of sdAb K2 in DC-based immunotherapy strategies.
Collapse
|
20
|
Evaluating a Single Domain Antibody Targeting Human PD-L1 as a Nuclear Imaging and Therapeutic Agent. Cancers (Basel) 2019; 11:cancers11060872. [PMID: 31234464 PMCID: PMC6628009 DOI: 10.3390/cancers11060872] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/13/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022] Open
Abstract
The PD-1:PD-L1 immune checkpoint axis is central in the escape of cancer cells from anticancer immune responses. Monoclonal antibodies (mAbs) specific for PD-L1 have been approved for treatment of various cancer types. Although PD-L1 blockade has proven its merit, there are still several aspects that require further attention to fully capitalize on its potential. One of these is the development of antigen-binding moieties that enable PD-L1 diagnosis and therapy. We generated human PD-L1 binding single domain antibodies (sdAbs) and selected sdAb K2, a sdAb with a high affinity for PD-L1, as a lead compound. SPECT/CT imaging in mice following intravenous injection of Technetium-99m (99mTc)-labeled sdAb K2 revealed high signal-to-noise ratios, strong ability to specifically detect PD-L1 in melanoma and breast tumors, and relatively low kidney retention, which is a unique property for radiolabeled sdAbs. We further showed using surface plasmon resonance that sdAb K2 binds to the same epitope on PD-L1 as the mAb avelumab, and antagonizes PD-1:PD-L1 interactions. Different human cell-based assays corroborated the PD-1:PD-L1 blocking activity, showing enhanced T-cell receptor signaling and tumor cell killing when PD-1POS T cells interacted with PD-L1POS tumor cells. Taken together, we present sdAb K2, which specifically binds to human PD-L1, as a new diagnostic and therapeutic agent in cancer management.
Collapse
|
21
|
Perforin and Granzyme B Expressed by Murine Myeloid-Derived Suppressor Cells: A Study on Their Role in Outgrowth of Cancer Cells. Cancers (Basel) 2019; 11:cancers11060808. [PMID: 31212684 PMCID: PMC6627828 DOI: 10.3390/cancers11060808] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/10/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
A wide-range of myeloid-derived suppressor cell (MDSC)-mediated immune suppressive functions has previously been described. Nevertheless, potential novel mechanisms by which MDSCs aid tumor progression are, in all likelihood, still unrecognized. Next to its well-known expression in natural killer cells and cytotoxic T lymphocytes (CTLs), granzyme B (GzmB) expression has been found in different cell types. In an MDSC culture model, we demonstrated perforin and GzmB expression. Furthermore, similar observations were made in MDSCs isolated from tumor-bearing mice. Even in MDSCs from humans, GzmB expression was demonstrated. Of note, B16F10 melanoma cells co-cultured with perforin/GzmB knock out mice (KO) MDSCs displayed a remarkable decrease in invasive potential. B16F10 melanoma cells co-injected with KO MDSCs, displayed a significant slower growth curve compared to tumor cells co-injected with wild type (WT) MDSCs. In vivo absence of perforin/GzmB in MDSCs resulted in a higher number of CD8+ T-cells. Despite this change in favor of CD8+ T-cell infiltration, we observed low interferon-γ (IFN-γ) and high programmed death-ligand 1 (PD-L1) expression, suggesting that other immunosuppressive mechanisms render these CD8+ T-cells dysfunctional. Taken together, our results suggest that GzmB expression in MDSCs is another means to promote tumor growth and warrants further investigation to unravel the exact underlying mechanism.
Collapse
|
22
|
De Vlaeminck Y, Lecocq Q, Giron P, Heirman C, Geeraerts X, Bolli E, Movahedi K, Massa S, Schoonooghe S, Thielemans K, Goyvaerts C, Van Ginderachter JA, Breckpot K. Single-domain antibody fusion proteins can target and shuttle functional proteins into macrophage mannose receptor expressing macrophages. J Control Release 2019; 299:107-120. [PMID: 30797866 DOI: 10.1016/j.jconrel.2019.02.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/20/2019] [Accepted: 02/18/2019] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment of numerous prevalent cancer types is abundantly infiltrated with tumor-associated macrophages (TAMs). Macrophage mannose receptor (MMR or CD206) expressing TAMs have been shown to be key promoters of tumor progression and major opponents of successful cancer therapy. Therefore, depleting MMR+ TAMs is an interesting approach to synergize with current antitumor therapies. We studied the potential of single-domain antibodies (sdAbs) specific for MMR to target proteins to MMR+ TAMs. Anti-MMR sdAbs were genetically coupled to a reporter protein, mWasabi (wasabi green, WG), generating sdAb "drug" fusion proteins (SFPs), referred to as WG-SFPs. The resulting WG-SFPs were highly efficient in targeting MMR+ macrophages both in vitro and in vivo. As we showed that second mitochondria-derived activator of caspase (SMAC) mimetics modulate MMR+ macrophages, we further coupled the anti-MMR sdAb to an active form of SMAC, referred to as tSMAC. The resulting tSMAC-SFPs were able to bind and upregulate caspase3/7 activity in MMR+ macrophages in vitro. In conclusion, we report the proof-of-concept of an elegant approach to conjugate anti-MMR sdAbs to proteins, which opens new avenues for targeted manipulation of MMR+ tumor-promoting TAMs.
Collapse
Affiliation(s)
- Yannick De Vlaeminck
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Quentin Lecocq
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Philippe Giron
- Laboratory of Medical and Molecular Oncology, Oncologic Research Centre, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Xenia Geeraerts
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Evangelia Bolli
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kiavash Movahedi
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sam Massa
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Steve Schoonooghe
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
23
|
De Beck L, Melhaoui S, De Veirman K, Menu E, De Bruyne E, Vanderkerken K, Breckpot K, Maes K. Epigenetic treatment of multiple myeloma mediates tumor intrinsic and extrinsic immunomodulatory effects. Oncoimmunology 2018; 7:e1484981. [PMID: 30288346 PMCID: PMC6169579 DOI: 10.1080/2162402x.2018.1484981] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 12/26/2022] Open
Abstract
Immune evasion is an important driver of disease progression in the plasma cell malignancy multiple myeloma. Recent work highlights the potential of epigenetic modulating agents as tool to enhance anti-tumor immunity. The immune modulating effects of the combination of a DNA methyltransferase inhibitor and a histone deacetylase inhibitor in multiple myeloma is insufficiently characterized. Therefore, we used the murine immunocompetent 5T33MM model to investigate hallmarks of immunogenic cell death as well as alterations in the immune cell constitution in the bone marrow of diseased mice in response to the DNA methyltransferase inhibitor decitabine and the histone deacetylase inhibitor quisinostat. Vaccination of mice with 5T33 cells treated with epigenetic compounds delayed tumor development upon a subsequent tumor challenge. In vitro, epigenetic treatment induced ecto-calreticulin and CD47, as well as a type I interferon response. Moreover, treated 5T33vt cells triggered dendritic cell maturation. The combination of decitabine and quisinostat in vivo resulted in combinatory anti-myeloma effects. In vivo, epigenetic treatment increased tumoral ecto-calreticulin and decreased CD47 and PD-L1 expression, increased dendritic cell maturation and reduced CD11b positive cells. Moreover, epigenetic treatment induced a temporal increase in presence of CD8-positive and CD4-positive T cells with naive and memory-like phenotypes based on CD62L and CD44 expression levels, and reduced expression of exhaustion markers PD-1 and TIM3. In conclusion, a combination of a DNA methyltransferase inhibitor and a histone deacetylase inhibitor increased the immunogenicity of myeloma cells and altered the immune cell constitution in the bone marrow of myeloma-bearing mice.
Collapse
Affiliation(s)
- Lien De Beck
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, Belgium.,Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sarah Melhaoui
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, Belgium
| |
Collapse
|
24
|
Versteven M, Van den Bergh JMJ, Broos K, Fujiki F, Campillo-Davo D, De Reu H, Morimoto S, Lecocq Q, Keyaerts M, Berneman Z, Sugiyama H, Van Tendeloo VFI, Breckpot K, Lion E. A versatile T cell-based assay to assess therapeutic antigen-specific PD-1-targeted approaches. Oncotarget 2018; 9:27797-27808. [PMID: 29963238 PMCID: PMC6021243 DOI: 10.18632/oncotarget.25591] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/24/2018] [Indexed: 12/27/2022] Open
Abstract
Blockade of programmed cell death protein 1 (PD-1) immune checkpoint receptor signaling is an established standard treatment for many types of cancer and indications are expanding. Successful clinical trials using monoclonal antibodies targeting PD-1 signaling have boosted preclinical research, encouraging development of novel therapeutics. Standardized assays to evaluate their bioactivity, however, remain restricted. The robust bioassays available all lack antigen-specificity. Here, we developed an antigen-specific, short-term and high-throughput T cell assay with versatile readout possibilities. A genetically modified T cell receptor (TCR)-deficient T cell line was stably transduced with PD-1. Transfection with messenger RNA encoding a TCR of interest and subsequent overnight stimulation with antigen-presenting cells, results in eGFP-positive and granzyme B-producing T cells for single cell or bulk analysis. Control antigen-presenting cells induced reproducible high antigen-specific eGFP and granzyme B expression. Upon PD-1 interaction, ligand-positive antigen-presenting immune or tumor cells elicited significantly lower eGFP and granzyme B expression, which could be restored by anti-PD-(L)1 blocking antibodies. This convenient cell-based assay shows a valuable tool for translational and clinical research on antigen-specific checkpoint-targeted therapy approaches.
Collapse
Affiliation(s)
- Maarten Versteven
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Johan M J Van den Bergh
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Katrijn Broos
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Soyoko Morimoto
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Division of Hematology, University Hospital Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Antwerp, Belgium
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Viggo F I Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Antwerp, Belgium
| |
Collapse
|
25
|
Non-invasive assessment of murine PD-L1 levels in syngeneic tumor models by nuclear imaging with nanobody tracers. Oncotarget 2018; 8:41932-41946. [PMID: 28410210 PMCID: PMC5522039 DOI: 10.18632/oncotarget.16708] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 03/19/2017] [Indexed: 12/31/2022] Open
Abstract
Blockade of the inhibitory PD-1/PD-L1 immune checkpoint axis is a promising cancer treatment. Nonetheless, a significant number of patients and malignancies do not respond to this therapy. To develop a screen for response to PD-1/PD-L1 inhibition, it is critical to develop a non-invasive tool to accurately assess dynamic immune checkpoint expression. Here we evaluated non-invasive SPECT/CT imaging of PD-L1 expression, in murine tumor models with varying PD-L1 expression, using high affinity PD-L1-specific nanobodies (Nbs). We generated and characterized 37 Nbs recognizing mouse PD-L1. Among those, four Nbs C3, C7, E2 and E4 were selected and evaluated for preclinical imaging of PD-L1 in syngeneic mice. We performed SPECT/CT imaging in wild type versus PD-L1 knock-out mice, using Technetium-99m (99mTc) labeled Nbs. Nb C3 and E2 showed specific antigen binding and beneficial biodistribution. Through the use of CRISPR/Cas9 PD-L1 knock-out TC-1 lung epithelial cell lines, we demonstrate that SPECT/CT imaging using Nb C3 and E2 identifies PD-L1 expressing tumors, but not PD-L1 non-expressing tumors, thereby confirming the diagnostic potential of the selected Nbs. In conclusion, these data show that Nbs C3 and E2 can be used to non-invasively image PD-L1 levels in the tumor, with the strength of the signal correlating with PD-L1 levels. These findings warrant further research into the use of Nbs as a tool to image inhibitory signals in the tumor environment.
Collapse
|
26
|
Pollack SM. The potential of the CMB305 vaccine regimen to target NY-ESO-1 and improve outcomes for synovial sarcoma and myxoid/round cell liposarcoma patients. Expert Rev Vaccines 2018; 17:107-114. [PMID: 29280411 PMCID: PMC6521962 DOI: 10.1080/14760584.2018.1419068] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Synovial Sarcoma (SS) and Myxoid Round Cell Liposarcoma (MRCL) are devastating sarcoma subtypes with few treatment options and poor outcomes in the advanced setting. However, both these diseases may be ideal for novel immunotherapies targeting the cancer-testis antigen, NY-ESO-1. AREAS COVERED In this review, we discuss the novel NY-ESO-1 targeted vaccine regimen, CMB305. This regimen uses a unique integration-deficient, dendritic-cell targeting lentiviral vector from the ZVex® platform, LV305, in order to prime NY-ESO-1 specific T cells. LV305 has single agent activity, and, in one case, caused a durable partial response in a refractory SS patient. CMB305 also includes a boost from a NY-ESO-1 protein vaccine given along with a potent toll-like-4 receptor agonist, glycopyranosyl lipid A. CMB305 induces NY-ESO-1 specific T cell responses in both SS and MRC patients and these patients had excellent overall survival (OS) outcomes in the initial phase I study. EXPERT COMMENTARY CMB305 is a therapeutic vaccine regimen targeting NY-ESO-1 based on the lentiviral vaccine vector, LV305. Phase I studies have proven this vaccine is active immunologically. Data suggesting this vaccine may improve OS for SS and MRCL patients is exciting but early, and on-going work is testing the impact of CMB305 on patient outcomes.
Collapse
Affiliation(s)
- Seth M Pollack
- a Clinical Research Division , Fred Hutchinson Cancer Research Center , Seattle , WA , USA
- b Department of Medicine , University of Washington , Seattle , WA , USA
| |
Collapse
|
27
|
Antigen-presenting cell-targeted lentiviral vectors do not support the development of productive T-cell effector responses: implications for in vivo targeted vaccine delivery. Gene Ther 2017; 24:370-375. [PMID: 28540936 DOI: 10.1038/gt.2017.30] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/24/2017] [Accepted: 04/10/2017] [Indexed: 12/22/2022]
Abstract
Targeting transgene expression specifically to antigen-presenting cells (APCs) has been put forward as a promising strategy to direct the immune system towards immunity. We developed the nanobody-display technology to restrict the tropism of lentiviral vectors (LVs) to APCs. However, we observed that immunization with APC-targeted LVs (DC2.1-LVs) did not evoke strong antigen-specific T-cell immunity when compared to immunization with broad tropism LVs (VSV.G-LVs). In this study, we report that VSV.G-LVs are more immunogenic than DC2.1-LVs because they transduce stromal cells, which has a role in activating antigen-specific T cells. Moreover, VSV.G-LVs trigger a pro-inflammatory innate immune response through transduction of APCs and stromal cells, while DC2.1-LVs trigger a type I interferon response with anti-viral capacity. These findings question the rationale of targeting LVs to APCs and argue for the development of VSV.G-LVs with an improved safety profile.
Collapse
|
28
|
Moyes KW, Lieberman NAP, Kreuser SA, Chinn H, Winter C, Deutsch G, Hoglund V, Watson R, Crane CA. Genetically Engineered Macrophages: A Potential Platform for Cancer Immunotherapy. Hum Gene Ther 2016; 28:200-215. [PMID: 27758144 DOI: 10.1089/hum.2016.060] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
In spite of their successes against hematologic malignancies, immunotherapeutic interventions for the treatment of patients with glioblastoma (GBM) have thus far been unsuccessful. This is in part due to the presence of a tumor microenvironment that fosters neoplastic growth and protects the tumor from destruction by the immune system. A novel genetically engineered macrophage-based platform has been developed with the potential to minimize the effects of the suppressive tumor microenvironment and improve innate and adaptive antitumor immune responses. A newly described lentiviral expression system was validated for the generation of transduced monocytes and monocyte-derived macrophages, and transgene expression was shown to be stable over the course of weeks to months, both in vitro and in a mouse xenograft model of GBM. Furthermore, the genetically engineered macrophages (GEMs) neither caused morbidity in animals nor contributed to accelerated tumor growth. The versatility of GEMs is also highlighted by showing that they can be engineered to secrete proteins that either reduce immune suppression, such as the soluble transforming growth factor beta receptor II, or promote immune cell activation, by expressing interleukin 21. There is also the potential to prevent GEM-mediated immune suppression by using the CRISPR system to knock out genes responsible for dysfunction of cytotoxic cells, including interleukin 10 and programmed death-ligand 1. Together, these results suggest that GEMs are an ideal cell type for transforming the tumor microenvironment and enhancing antitumor immunity. Importantly, it is anticipated that these findings will have broad applicability to other types of tumors with microenvironments that currently preclude successful immunotherapeutic approaches.
Collapse
Affiliation(s)
- Kara W Moyes
- 1 Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute , Seattle, Washington
| | - Nicole A P Lieberman
- 1 Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute , Seattle, Washington
| | - Shannon A Kreuser
- 1 Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute , Seattle, Washington
| | - Harrison Chinn
- 1 Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute , Seattle, Washington
| | - Conrad Winter
- 2 Department of Pathology, Seattle Children's Hospital, Seattle, Washington
| | - Gail Deutsch
- 2 Department of Pathology, Seattle Children's Hospital, Seattle, Washington
| | - Virginia Hoglund
- 1 Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute , Seattle, Washington
| | - Reid Watson
- 1 Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute , Seattle, Washington
| | - Courtney A Crane
- 1 Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute , Seattle, Washington
| |
Collapse
|
29
|
Abstract
Adoptive cellular therapy represents a robust means of augmenting the tumor-reactive effector population in patients with cancer by adoptive transfer of ex vivo expanded T cells. Three approaches have been developed to achieve this goal: the use of tumor-infiltrating lymphocytes or tumor-infiltrating lymphocytess extracted from patient biopsy material; the redirected engineering of lymphocytes using vectors expressing a chimeric antigen receptor and T-cell receptor; and third, the isolation and expansion of often low-frequency endogenous T cells (ETCs) reactive to tumor antigens from the peripheral blood of patients. This last form of adoptive transfer of T cells, known as ETC therapy, requires specialized methods to isolate and expand from peripheral blood the very low-frequency tumor-reactive T cells, methods that have been developed over the last 2 decades, to the point where such an approach may be broadly applicable not only for the treatment of melanoma but also for that of other solid tumor malignancies. One compelling feature of ETC is the ability to rapidly deploy clinical trials following identification of a tumor-associated target epitope, a feature that may be exploited to develop personalized antigen-specific T-cell therapy for patients with almost any solid tumor. With a well-validated antigen discovery pipeline in place, clinical studies combining ETC with agents that modulate the immune microenvironment can be developed that will transform ETC into a feasible treatment modality.
Collapse
|
30
|
Dufait I, Van Valckenborgh E, Menu E, Escors D, De Ridder M, Breckpot K. Signal transducer and activator of transcription 3 in myeloid-derived suppressor cells: an opportunity for cancer therapy. Oncotarget 2016; 7:42698-42715. [PMID: 27029037 PMCID: PMC5173167 DOI: 10.18632/oncotarget.8311] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/23/2016] [Indexed: 12/13/2022] Open
Abstract
Cancer progression is in part determined by interactions between cancer cells and stromal cells in the tumor microenvironment (TME). The identification of cytotoxic tumor-infiltrating lymphocytes has instigated research into immune stimulating cancer therapies. Although a promising direction, immunosuppressive mechanisms exerted at the TME hamper its success. Myeloid-derived suppressor cells (MDSCs) have come to the forefront as stromal cells that orchestrate the immunosuppressive TME. Consequently, this heterogeneous cell population has been the object of investigation. Studies revealed that the transcription factor signal transducer and activator of transcription 3 (STAT3) largely dictates the recruitment, activation and function of MDSCs in the TME. Therefore, this review will focus on the role of this key transcription factor during the MDSC's life cycle and on the therapeutic opportunities it offers.
Collapse
Affiliation(s)
- Inès Dufait
- Department of Radiotherapy, Vrije Universiteit, UZ-Brussel, Brussels, Belgium
- Laboratory of Molecular and Cellular Technology, Vrije Universiteit, UZ-Brussel, Brussels, Belgium
| | - Els Van Valckenborgh
- Laboratory of Hematology and Immunology, Vrije Universiteit, UZ-Brussel, Brussels, Belgium
| | - Eline Menu
- Laboratory of Hematology and Immunology, Vrije Universiteit, UZ-Brussel, Brussels, Belgium
| | - David Escors
- Immunomodulation Group, Navarrabiomed-Fundaçion, Miguel Servet, IdiSNA, Navarra, Spain
| | - Mark De Ridder
- Department of Radiotherapy, Vrije Universiteit, UZ-Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Technology, Vrije Universiteit, UZ-Brussel, Brussels, Belgium
| |
Collapse
|
31
|
Tosatto A, Sommaggio R, Kummerow C, Bentham RB, Blacker TS, Berecz T, Duchen MR, Rosato A, Bogeski I, Szabadkai G, Rizzuto R, Mammucari C. The mitochondrial calcium uniporter regulates breast cancer progression via HIF-1α. EMBO Mol Med 2016; 8:569-85. [PMID: 27138568 PMCID: PMC4864890 DOI: 10.15252/emmm.201606255] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 12/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) represents the most aggressive breast tumor subtype. However, the molecular determinants responsible for the metastatic TNBC phenotype are only partially understood. We here show that expression of the mitochondrial calcium uniporter (MCU), the selective channel responsible for mitochondrial Ca(2+) uptake, correlates with tumor size and lymph node infiltration, suggesting that mitochondrial Ca(2+) uptake might be instrumental for tumor growth and metastatic formation. Accordingly, MCU downregulation hampered cell motility and invasiveness and reduced tumor growth, lymph node infiltration, and lung metastasis in TNBC xenografts. In MCU-silenced cells, production of mitochondrial reactive oxygen species (mROS) is blunted and expression of the hypoxia-inducible factor-1α (HIF-1α) is reduced, suggesting a signaling role for mROS and HIF-1α, downstream of mitochondrial Ca(2+) Finally, in breast cancer mRNA samples, a positive correlation of MCU expression with HIF-1α signaling route is present. Our results indicate that MCU plays a central role in TNBC growth and metastasis formation and suggest that mitochondrial Ca(2+) uptake is a potential novel therapeutic target for clinical intervention.
Collapse
Affiliation(s)
- Anna Tosatto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Roberta Sommaggio
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Carsten Kummerow
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Robert B Bentham
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | - Thomas S Blacker
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | - Tunde Berecz
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | - Michael R Duchen
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Ivan Bogeski
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Gyorgy Szabadkai
- Department of Biomedical Sciences, University of Padua, Padua, Italy Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy CNR Institute of Neuroscience, National Council of Research, Padua, Italy
| | | |
Collapse
|
32
|
Dufait I, Schwarze JK, Liechtenstein T, Leonard W, Jiang H, Escors D, De Ridder M, Breckpot K. Ex vivo generation of myeloid-derived suppressor cells that model the tumor immunosuppressive environment in colorectal cancer. Oncotarget 2016; 6:12369-82. [PMID: 25869209 PMCID: PMC4494944 DOI: 10.18632/oncotarget.3682] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 03/11/2015] [Indexed: 12/27/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of cells that accumulate in tumor-bearing subjects and which strongly inhibit anti-cancer immune responses. To study the biology of MDSC in colorectal cancer (CRC), we cultured bone marrow cells in conditioned medium from CT26 cells, which are genetically modified to secrete high levels of granulocyte-macrophage colony-stimulating factor. This resulted in the generation of high numbers of CD11b(+) Ly6G(+) granulocytic and CD11b(+) Ly6C(+) monocytic MDSC, which closely resemble those found within the tumor but not the spleen of CT26 tumor-bearing mice. Such MDSC potently inhibited T-cell responses in vitro, a process that could be reversed upon blocking of arginase-1 or inducible nitric oxide synthase (iNOS). We confirmed that inhibition of arginase-1 or iNOS in vivo resulted in the stimulation of cytotoxic T-cell responses. A delay in tumor growth was observed upon functional repression of both enzymes. These data confirm the role of MDSC as inhibitors of T-cell-mediated immune responses in CRC. Moreover, MDSC differentiated in vitro from bone marrow cells using conditioned medium of GM-CSF-secreting CT26 cells, represent a valuable platform to study/identify drugs that counteract MDSC activities.
Collapse
Affiliation(s)
- Inès Dufait
- UZ Brussel, Department of Radiotherapy, Vrije Universiteit Brussel, Brussels, Belgium.,Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Therese Liechtenstein
- Navarrabiomed-Fundaçion Miguel Servet, Immunomodulation Group, Pamplona, Spain.,Division of Infection and Immunity, University College London, London, UK
| | - Wim Leonard
- UZ Brussel, Department of Radiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Heng Jiang
- UZ Brussel, Department of Radiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - David Escors
- Navarrabiomed-Fundaçion Miguel Servet, Immunomodulation Group, Pamplona, Spain.,Division of Infection and Immunity, University College London, London, UK
| | - Mark De Ridder
- UZ Brussel, Department of Radiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
33
|
Abstract
Basic science advances in cancer immunotherapy have resulted in various treatments that have recently shown success in the clinic. Many of these therapies require the insertion of genes into cells to directly kill them or to redirect the host's cells to induce potent immune responses. Other analogous therapies work by modifying effector cells for improved targeting and enhanced killing of tumor cells. Initial studies done using γ-retroviruses were promising, but safety concerns centered on the potential for insertional mutagenesis have highlighted the desire to develop other options for gene delivery. Lentiviral vectors (LVs) have been identified as potentially more effective and safer alternative delivery vehicles. LVs are now in use in clinical trials for many different types of inherited and acquired disorders, including cancer. This review will discuss current knowledge of LVs and the applications of this viral vector-based delivery vehicle to cancer immunotherapy.
Collapse
Affiliation(s)
- Robyn Aa Oldham
- Department of Medical Biophysics, University of Toronto, 27 King's College Circle, Toronto, ON M5S, Canada
| | | | | |
Collapse
|
34
|
Van Lint S, Renmans D, Broos K, Goethals L, Maenhout S, Benteyn D, Goyvaerts C, Du Four S, Van der Jeught K, Bialkowski L, Flamand V, Heirman C, Thielemans K, Breckpot K. Intratumoral Delivery of TriMix mRNA Results in T-cell Activation by Cross-Presenting Dendritic Cells. Cancer Immunol Res 2015; 4:146-56. [DOI: 10.1158/2326-6066.cir-15-0163] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 10/23/2015] [Indexed: 01/02/2023]
|
35
|
Dai S, Zhuo M, Song L, Chen X, Yu Y, Tang Z, Zang G. Dendritic cell-based vaccination with lentiviral vectors encoding ubiquitinated hepatitis B core antigen enhances hepatitis B virus-specific immune responses in vivo. Acta Biochim Biophys Sin (Shanghai) 2015; 47:870-9. [PMID: 26373843 DOI: 10.1093/abbs/gmv093] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/30/2015] [Indexed: 12/27/2022] Open
Abstract
The activity of hepatitis B virus (HBV)-specific cytotoxic T lymphocytes (CTLs) plays a predominant role in the clearance of HBV. Dendritic cells (DCs) are key antigen-presenting cells and play an important role in the initiation of immune responses. We previously verified that lentiviral vector encoding ubiquitinated hepatitis B core antigen (LV-Ub-HBcAg) effectively transduced DCs to induce maturation, and the mature DCs efficiently induced T cell polarization to Th1 and generated HBcAg-specific CTLs ex vivo. In this study, HBV-specific immune responses of LV-Ub-HBcAg in BALB/c mice (H-2Kd) were evaluated. It was shown that direct injection of LV-Ub-HBcAg increased the production of cytokines IL-2 and IFN-γ, elicited strong antibody responses, and remarkably generated a high percentage of IFN-γ+CD8+ T cells with HBV-specific CTL responses in BALB/c mice. In addition, direct injection of LV-Ub-HBcAg induced potent anti-HBV immune responses, similar to those elicited by in vitro-transduced DCs. In conclusion, the DC-based therapeutic vaccine LV-Ub-HBcAg elicited specific antibody immune responses and induced robust specific CTL activity in vivo.
Collapse
Affiliation(s)
- Shenglan Dai
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Meng Zhuo
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Linlin Song
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xiaohua Chen
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yongsheng Yu
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zhenghao Tang
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Guoqing Zang
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
36
|
Mullins CS, Wegner T, Klar E, Classen CF, Linnebacher M. Optimizing the process of nucleofection for professional antigen presenting cells. BMC Res Notes 2015; 8:472. [PMID: 26404473 PMCID: PMC4581479 DOI: 10.1186/s13104-015-1446-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/15/2015] [Indexed: 12/23/2022] Open
Abstract
Background In times of rapidly increasing numbers of immunological approaches entering the clinics, antigen delivery becomes a pivotal process. The genuine way of rendering antigen presenting cells (APC) antigen specific, largely influences the outcome of the immune response. Short peptides bear the demerit of HLA restriction, whereas the proper way of delivery for long peptide sequences is currently a matter of debate. Electroporation is a reliable method for antigen delivery, especially using nucleic acids. The nucleofection process is based on this approach with the twist of further ensuring delivery also into the nucleus. Beside the form of antigen, the type of APC used for immune response induction may be crucial. Dendritic cells (DC) are by far the most commonly used APC; however B cells have entered this field as well and have gained wide acceptance. Results In this study, we compared B cells to DC with regard to nucleofection efficiency and intensity of resulting antigen expression. APC were transfected either with plasmid DNA containing the reporter gene green fluorescent protein (GFP) or directly with in vitro-transcribed (IVT) GPF mRNA as a surrogate antigen. Out of nearly 100 different nucleofection programs tested, the top five for each cell type were identified and validated using cells from cancer patients. Flow cytometric analyses of transfected cells determining GFP expression and viability revealed a reverse correlation of efficiency and viability. Finally, donor dependant variances were analyzed. Conclusion In summary, nucleofection of both DC and B cells is feasible with plasmid DNA and IVT mRNA. And no differences with regard to nucleofectability were observed between the two cell types. Using IVT mRNA omits the danger of genomic integration and plasmid DNA constructs permit a more potent and longer lasting antigen expression. Electronic supplementary material The online version of this article (doi:10.1186/s13104-015-1446-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christina Susanne Mullins
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Hospital Rostock, Schillingallee 35, 18057, Rostock, Germany. .,University Children's Hospital Rostock, Ernst-Heydemann-Str. 8, 18057, Rostock, Germany.
| | - Tabea Wegner
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Hospital Rostock, Schillingallee 35, 18057, Rostock, Germany.
| | - Ernst Klar
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Hospital Rostock, Schillingallee 35, 18057, Rostock, Germany.
| | - Carl-Friedrich Classen
- University Children's Hospital Rostock, Ernst-Heydemann-Str. 8, 18057, Rostock, Germany.
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Hospital Rostock, Schillingallee 35, 18057, Rostock, Germany.
| |
Collapse
|
37
|
Amodio G, Annoni A, Gregori S. Dendritic Cell Immune Therapy to Break or Induce Tolerance. CURRENT STEM CELL REPORTS 2015. [DOI: 10.1007/s40778-015-0024-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
38
|
Specific inhibition of mTOR pathway induces anti-proliferative effect and decreases the hormone secretion in cultured pituitary adenoma cells. J Neurooncol 2015; 125:79-89. [DOI: 10.1007/s11060-015-1895-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 08/14/2015] [Indexed: 02/02/2023]
|
39
|
Van der Jeught K, Joe PT, Bialkowski L, Heirman C, Daszkiewicz L, Liechtenstein T, Escors D, Thielemans K, Breckpot K. Intratumoral administration of mRNA encoding a fusokine consisting of IFN-β and the ectodomain of the TGF-β receptor II potentiates antitumor immunity. Oncotarget 2015; 5:10100-13. [PMID: 25338019 PMCID: PMC4259408 DOI: 10.18632/oncotarget.2463] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 09/06/2014] [Indexed: 12/14/2022] Open
Abstract
It is generally accepted that the success of immunotherapy depends on the presence of tumor-specific CD8⁺ cytotoxic T cells and the modulation of the tumor environment. In this study, we validated mRNA encoding soluble factors as a tool to modulate the tumor microenvironment to potentiate infiltration of tumor-specific T cells. Intratumoral delivery of mRNA encoding a fusion protein consisting of interferon-β and the ectodomain of the transforming growth factor-β receptor II, referred to as Fβ², showed therapeutic potential. The treatment efficacy was dependent on CD8⁺ T cells and could be improved through blockade of PD-1/PD-L1 interactions. In vitro studies revealed that administration of Fβ² to tumor cells resulted in a reduced proliferation and increased expression of MHC I but also PD-L1. Importantly, Fβ² enhanced the antigen presenting capacity of dendritic cells, whilst reducing the suppressive activity of myeloid-derived suppressor cells. In conclusion, these data suggest that intratumoral delivery of mRNA encoding soluble proteins, such as Fβ², can modulate the tumor microenvironment, leading to effective antitumor T cell responses, which can be further potentiated through combination therapy.
Collapse
Affiliation(s)
- Kevin Van der Jeught
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Patrick Tjok Joe
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lukasz Bialkowski
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lidia Daszkiewicz
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - David Escors
- Rayne Institute, University College London, London, UK. Biomedical Research Centre NavarraBiomed-Fundacion Miguel Servet, National Health Service of Navarre, Pamplona, Navarre, Spain
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
40
|
Filatenkov A, Baker J, Mueller AMS, Kenkel J, Ahn GO, Dutt S, Zhang N, Kohrt H, Jensen K, Dejbakhsh-Jones S, Shizuru JA, Negrin RN, Engleman EG, Strober S. Ablative Tumor Radiation Can Change the Tumor Immune Cell Microenvironment to Induce Durable Complete Remissions. Clin Cancer Res 2015; 21:3727-39. [PMID: 25869387 DOI: 10.1158/1078-0432.ccr-14-2824] [Citation(s) in RCA: 350] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/15/2015] [Indexed: 01/19/2023]
Abstract
PURPOSE The goals of the study were to elucidate the immune mechanisms that contribute to desirable complete remissions of murine colon tumors treated with single radiation dose of 30 Gy. This dose is at the upper end of the ablative range used clinically to treat advanced or metastatic colorectal, liver, and non-small cell lung tumors. EXPERIMENTAL DESIGN Changes in the tumor immune microenvironment of single tumor nodules exposed to radiation were studied using 21-day (>1 cm in diameter) CT26 and MC38 colon tumors. These are well-characterized weakly immunogenic tumors. RESULTS We found that the high-dose radiation transformed the immunosuppressive tumor microenvironment resulting in an intense CD8(+) T-cell tumor infiltrate, and a loss of myeloid-derived suppressor cells (MDSC). The change was dependent on antigen cross-presenting CD8(+) dendritic cells, secretion of IFNγ, and CD4(+)T cells expressing CD40L. Antitumor CD8(+) T cells entered tumors shortly after radiotherapy, reversed MDSC infiltration, and mediated durable remissions in an IFNγ-dependent manner. Interestingly, extended fractionated radiation regimen did not result in robust CD8(+) T-cell infiltration. CONCLUSIONS For immunologically sensitive tumors, these results indicate that remissions induced by a short course of high-dose radiotherapy depend on the development of antitumor immunity that is reflected by the nature and kinetics of changes induced in the tumor cell microenvironment. These results suggest that systematic examination of the tumor immune microenvironment may help in optimizing the radiation regimen used to treat tumors by adding a robust immune response.
Collapse
Affiliation(s)
- Alexander Filatenkov
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California.
| | - Jeanette Baker
- Division of Blood and Bone Marrow Transplantation, Department of Medicine, Stanford University, School of Medicine, Stanford, California
| | - Antonia M S Mueller
- Division of Blood and Bone Marrow Transplantation, Department of Medicine, Stanford University, School of Medicine, Stanford, California
| | - Justin Kenkel
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - G-One Ahn
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Suparna Dutt
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Nigel Zhang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Holbrook Kohrt
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Kent Jensen
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Sussan Dejbakhsh-Jones
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Judith A Shizuru
- Division of Blood and Bone Marrow Transplantation, Department of Medicine, Stanford University, School of Medicine, Stanford, California
| | - Robert N Negrin
- Division of Blood and Bone Marrow Transplantation, Department of Medicine, Stanford University, School of Medicine, Stanford, California
| | - Edgar G Engleman
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Samuel Strober
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
41
|
Julier Z, Martino MM, de Titta A, Jeanbart L, Hubbell JA. The TLR4 agonist fibronectin extra domain A is cryptic, exposed by elastase-2; use in a fibrin matrix cancer vaccine. Sci Rep 2015; 5:8569. [PMID: 25708982 PMCID: PMC4338432 DOI: 10.1038/srep08569] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/23/2015] [Indexed: 11/09/2022] Open
Abstract
Fibronectin (FN) is an extracellular matrix (ECM) protein including numerous fibronectin type III (FNIII) repeats with different functions. The alternatively spliced FN variant containing the extra domain A (FNIII EDA), located between FNIII 11 and FNIII 12, is expressed in sites of injury, chronic inflammation, and solid tumors. Although its function is not well understood, FNIII EDA is known to agonize Toll-like receptor 4 (TLR4). Here, by producing various FN fragments containing FNIII EDA, we found that FNIII EDA's immunological activity depends upon its local intramolecular context within the FN chain. N-terminal extension of the isolated FNIII EDA with its neighboring FNIII repeats (FNIII 9-10-11) enhanced its activity in agonizing TLR4, while C-terminal extension with the native FNIII 12-13-14 heparin-binding domain abrogated it. In addition, we reveal that an elastase 2 cleavage site is present between FNIII EDA and FNIII 12. Activity of the C-terminally extended FNIII EDA could be restored after cleavage of the FNIII 12-13-14 domain by elastase 2. FN being naturally bound to the ECM, we immobilized FNIII EDA-containing FN fragments within a fibrin matrix model along with antigenic peptides. Such matrices were shown to stimulate cytotoxic CD8+ T cell responses in two murine cancer models.
Collapse
Affiliation(s)
- Ziad Julier
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Mikaël M Martino
- 1] Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland [2] World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Alexandre de Titta
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Laura Jeanbart
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Jeffrey A Hubbell
- 1] Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland [2] Institute for Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland [3] Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA [4] Materials Science Division, Argonne National Laboratory, Argonne, IL 60439, USA
| |
Collapse
|
42
|
Li X, Gu S, Ling Y, Shen C, Cao X, Xie R. p53 inhibition provides a pivotal protective effect against ischemia-reperfusion injury in vitro via mTOR signaling. Brain Res 2015; 1605:31-8. [PMID: 25681550 DOI: 10.1016/j.brainres.2015.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 02/06/2023]
Abstract
Tumor suppressor p53 has recently been reported to have numerous functions independent of tumorigenesis, including neuronal survival during ischemia. The mammalian target of rapamycin (mTOR) signaling pathway plays a central role in the regulation of metabolism, cell growth, development, and cell survival. Our recent work has demonstrated the neuroprotective effects of the mTOR pathway. Considering that p53 is also an important regulator of mTOR, to further clarify the role of p53 and the mTOR signaling pathway in neuronal ischemic-reperfusion injury, we used mouse primary mixed cultured neurons with an oxygen glucose deprivation (OGD) model to mimic an ischemic-reperfusion injury in vitro. A lentiviral system was also used to inhibit or overexpress p53 to determine whether p53 alteration affects OGD and reperfusion injury. Our results show that activated p53 was induced and it suppressed mTOR expression in primary mixed cultured neurons after OGD and reperfusion. Inhibiting p53, using either a chemical inhibitor or lentiviral-mediated shRNA, exhibited neuroprotective effects in primary cultured neurons against OGD and reperfusion injury through the upregulation of mTOR activity. Such protective effects could be reversed by rapamycin, an mTOR inhibitor. Conversely, p53 overexpression tended to exacerbate the detrimental effects of OGD injury by downregulating mTOR activity. These results suggest that p53 inhibition has a pivotal protective effect against an in vitro ischemia-reperfusion injury via mTOR signaling and provides a potential and promising therapeutic target for stroke treatment.
Collapse
Affiliation(s)
- Xiaomu Li
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shixin Gu
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Yan Ling
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chao Shen
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Xiaoyun Cao
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Rong Xie
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China.
| |
Collapse
|
43
|
Van der Jeught K, Bialkowski L, Daszkiewicz L, Broos K, Goyvaerts C, Renmans D, Van Lint S, Heirman C, Thielemans K, Breckpot K. Targeting the tumor microenvironment to enhance antitumor immune responses. Oncotarget 2015; 6:1359-81. [PMID: 25682197 PMCID: PMC4359300 DOI: 10.18632/oncotarget.3204] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 12/24/2014] [Indexed: 12/16/2022] Open
Abstract
The identification of tumor-specific antigens and the immune responses directed against them has instigated the development of therapies to enhance antitumor immune responses. Most of these cancer immunotherapies are administered systemically rather than directly to tumors. Nonetheless, numerous studies have demonstrated that intratumoral therapy is an attractive approach, both for immunization and immunomodulation purposes. Injection, recruitment and/or activation of antigen-presenting cells in the tumor nest have been extensively studied as strategies to cross-prime immune responses. Moreover, delivery of stimulatory cytokines, blockade of inhibitory cytokines and immune checkpoint blockade have been explored to restore immunological fitness at the tumor site. These tumor-targeted therapies have the potential to induce systemic immunity without the toxicity that is often associated with systemic treatments. We review the most promising intratumoral immunotherapies, how these affect systemic antitumor immunity such that disseminated tumor cells are eliminated, and which approaches have been proven successful in animal models and patients.
Collapse
Affiliation(s)
- Kevin Van der Jeught
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Lukasz Bialkowski
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Lidia Daszkiewicz
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Katrijn Broos
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Cleo Goyvaerts
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Dries Renmans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Sandra Van Lint
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| |
Collapse
|
44
|
Goyvaerts C, Kurt DG, Van Lint S, Heirman C, Van Ginderachter JA, De Baetselier P, Raes G, Thielemans K, Breckpot K. Immunogenicity of targeted lentivectors. Oncotarget 2015; 5:704-15. [PMID: 24519916 PMCID: PMC3996667 DOI: 10.18632/oncotarget.1680] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To increase the safety and possibly efficacy of HIV-1 derived lentivectors (LVs) as an anti-cancer vaccine, we recently developed the Nanobody (Nb) display technology to target LVs to antigen presenting cells (APCs). In this study, we extend these data with exclusive targeting of LVs to conventional dendritic cells (DCs), which are believed to be the main cross-presenting APCs for the induction of a TH1-conducted antitumor immune response. The immunogenicity of these DC-subtype targeted LVs was compared to that of broad tropism, general APC-targeted and non-infectious LVs. Intranodal immunization with ovalbumin encoding LVs induced proliferation of antigen specific CD4+ T cells, irrespective of the LVs' targeting ability. However, the cytokine secretion profile of the restimulated CD4+ T cells demonstrated that general APC targeting induced a similar TH1-profile as the broad tropism LVs while transduction of conventional DCs alone induced a similar and less potent TH1 profile as the non-infectious LVs. This observation contradicts the hypothesis that conventional DCs are the most important APCs and suggests that the activation of other APCs is also meaningful. Despite these differences, all targeted LVs were able to stimulate cytotoxic T lymphocytes, be it to a lesser extent than broad tropism LVs. Furthermore this induction was shown to be dependent on type I interferon for the targeted and non-infectious LVs, but not for broad tropism LVs. Finally we demonstrated that the APC-targeted LVs were as potent in therapy as broad tropism LVs and as such deliver on their promise as safer and efficacious LV-based vaccines.
Collapse
Affiliation(s)
- Cleo Goyvaerts
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Silva JM, Zupancic E, Vandermeulen G, Oliveira VG, Salgado A, Videira M, Gaspar M, Graca L, Préat V, Florindo HF. In vivo delivery of peptides and Toll-like receptor ligands by mannose-functionalized polymeric nanoparticles induces prophylactic and therapeutic anti-tumor immune responses in a melanoma model. J Control Release 2014; 198:91-103. [PMID: 25483429 DOI: 10.1016/j.jconrel.2014.11.033] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/24/2014] [Accepted: 11/26/2014] [Indexed: 10/24/2022]
Abstract
We hypothesized that the co-entrapment of melanoma-associated antigens and the Toll-like receptor (TLR) ligands Poly(I:C) and CpG, known to be Th1-immunopotentiators, in mannose-functionalized aliphatic polyester-based nanoparticles (NPs) could be targeted to mannose receptors on antigen-presenting cells and induce anti-tumor immune responses. High entrapment efficiencies of antigens and immunopotentiators in 150nm NPs were obtained. The co-entrapment of the model antigen ovalbumin and the TLR ligands was crucial to induce high IgG2c/IgG1 ratios and high levels of IFN-γ and IL-2. Mannose-functionalization of NPs potentiated the Th1 immune response. The nanoparticulate vaccines decreased the growth rate of murine B16F10 melanoma tumors in therapeutic and prophylatic settings. The combination of mannose-functionalized NPs containing MHC class I- or class II-restricted melanoma antigens and the TLR ligands induced the highest tumor growth delay. Overall, we demonstrate that the multifunctional properties of NPs in terms of targeting and antigen/adjuvant delivery have high cancer immunotherapeutic potential.
Collapse
Affiliation(s)
- Joana M Silva
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal; Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Eva Zupancic
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Gaëlle Vandermeulen
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Vanessa G Oliveira
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-025 Lisbon, Portugal
| | - Ana Salgado
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Mafalda Videira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Manuela Gaspar
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Luis Graca
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-025 Lisbon, Portugal
| | - Véronique Préat
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université Catholique de Louvain, 1200 Brussels, Belgium.
| | - Helena F Florindo
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal.
| |
Collapse
|
46
|
Bieghs L, Lub S, Fostier K, Maes K, Van Valckenborgh E, Menu E, Johnsen HE, Overgaard MT, Larsson O, Axelson M, Nyegaard M, Schots R, Jernberg-Wiklund H, Vanderkerken K, De Bruyne E. The IGF-1 receptor inhibitor picropodophyllin potentiates the anti-myeloma activity of a BH3-mimetic. Oncotarget 2014; 5:11193-208. [PMID: 25008202 PMCID: PMC4294345 DOI: 10.18632/oncotarget.1933] [Citation(s) in RCA: 13] [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: 02/26/2014] [Accepted: 04/30/2014] [Indexed: 12/22/2022] Open
Abstract
The ABT-analogous 737, 263 and 199 are BH3 mimetics showing potent anti-myeloma (MM) activity, but only on defined molecular subgroups of MM patients presenting a Bcl-2high/Mcl-1low profile. IGF-1 is a major survival factor in MM regulating the expression of Bcl-2 proteins and might therefore be a resistance factor to these ABT-analogous. We first show that IGF-1 protected human MM cell lines (HMCLs) against ABT-737. Concurrently, the IGF-1 receptor inhibitor picropodophyllin (PPP) synergistically sensitized HMCL, primary human MM and murine 5T33MM cells to ABT-737 and ABT-199 by further decreasing cell viability and enhancing apoptosis. Knockdown of Bcl-2 by shRNA protected MM cells to ABT-737, while Mcl-1 shRNA sensitized the cells. PPP overcame the Bcl-2 dependency of ABT-737, but failed to completely overcome the protective effect of Mcl-1. In vivo, co-treatment of 5T33MM bearing mice significantly decreased tumor burden and prolonged overall survival both in a prophylactic and therapeutic setting. Interestingly, proteasome inhibitor resistant CD138- 5T33MM cells were more sensitive to ABT-737, whereas PPP alone targeted the CD138+ cells more effectively. After co-treatment, both subpopulations were targeted equally. Together, the combination of an IGF-1R inhibitor and an ABT-analogue displays synergistic anti-myeloma activity providing the rational for further (pre)clinical testing.
Collapse
Affiliation(s)
- Liesbeth Bieghs
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Haematology, Aalborg Hospital, Aalborg University, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Susanne Lub
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karel Fostier
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ken Maes
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Els Van Valckenborgh
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hans E. Johnsen
- Department of Haematology, Aalborg Hospital, Aalborg University, Denmark
| | | | - Olle Larsson
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Magnus Axelson
- Department of Clinical Chemistry, Karolinska Hospital, Stockholm, Sweden
| | - Mette Nyegaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Rik Schots
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Karin Vanderkerken
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
47
|
Abstract
Adoptive T-cell therapy involves the ex vivo enrichment and expansion of tumor-reactive T cells for infusion. As an immune-based approach, adoptive therapy has become an increasingly attractive modality for the treatment of patients with cancer due to its potential for high specificity, non-cross resistance with conventional therapies, and promise of long-term immunoprotection. In recent years, a resurgence in discoveries underlying T-cell recognition, tumor immune evasion, and T-cell memory and differentiation coupled with the development of several enabling technologies have facilitated a renewed focus in the field of adoptive therapy and its transition to the clinical arena as a treatment modality for patients with cancer. In this review, endogenous T cells derived from peripheral blood or tumor sites will be presented as a source of effector cells for adoptive therapy and strategies to isolate, manipulate, and enhance the function of antigen-specific T cells in vitro and to augment their in vivo efficacy and persistence by host immunomodulation are presented in the context of an ever-increasing inventory of preclinical and clinically available reagents. Optimizing the combination of adoptive cellular therapy and other immune-based and conventional approaches will herald a new generation of research and clinical opportunities for cancer immunotherapy.
Collapse
Affiliation(s)
- Cassian Yee
- Department of Melanoma Medical Oncology and Department of Immunology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
48
|
Filatenkov A, Baker J, Müller AM, Ahn GO, Kohrt H, Dutt S, Jensen K, Dejbakhsh-Jones S, Negrin RS, Shizuru JA, Engleman EG, Strober S. Treatment of 4T1 metastatic breast cancer with combined hypofractionated irradiation and autologous T-cell infusion. Radiat Res 2014; 182:163-9. [PMID: 24992165 DOI: 10.1667/rr13471.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The goal of this study was to determine whether a combination of local tumor irradiation and autologous T-cell transplantation can effectively treat metastatic 4T1 breast cancer in mice. BALB/c mice were injected subcutaneously with luciferase-labeled 4T1 breast tumor cells and allowed to grow for 21 days, at which time metastases appeared in the lungs. Primary tumors were treated at that time with 3 daily fractions of 20 Gy of radiation each. Although this approach could eradicate primary tumors, tumors in the lungs grew progressively. We attempted to improve efficacy of the radiation by adding autologous T-cell infusions. Accordingly, T cells were purified from the spleens of tumor-bearing mice after completion of irradiation and cryopreserved. Cyclophosphamide was administered thereafter to induce lymphodepletion, followed by T-cell infusion. Although the addition of cyclophosphamide to irradiation did not improve survival or reduce tumor progression, the combination of radiation, cyclophosphamide and autologous T-cell infusion induced durable remissions and markedly improved survival. We conclude that the combination of radiation and autologous T-cell infusion is an effective treatment for metastatic 4T1 breast cancer.
Collapse
Affiliation(s)
- Alexander Filatenkov
- a Division of Immunology and Rheumatology, Department of Medicine, Stanford University, School of Medicine, Stanford, California 94305
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kallen KJ, Theß A. A development that may evolve into a revolution in medicine: mRNA as the basis for novel, nucleotide-based vaccines and drugs. THERAPEUTIC ADVANCES IN VACCINES 2014; 2:10-31. [PMID: 24757523 DOI: 10.1177/2051013613508729] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent advances strongly suggest that mRNA rather than DNA will be the nucleotide basis for a new class of vaccines and drugs. Therapeutic cancer vaccines against a variety of targets have been developed on this basis and initial clinical experience suggests that preclinical activity can be successfully translated to human application. Likewise, prophylactic vaccines against viral pathogens and allergens have demonstrated their activity in animal models. These successes could be extended preclinically to mRNA protein and gene replacement therapy as well as the induction of pluripotent stem cells by mRNA encoded transcription factors. The production of mRNA-based vaccines and drugs is highly flexible, scalable and cost competitive, and eliminates the requirement of a cold chain. mRNA-based drugs and vaccines offer all the advantages of a nucleotide-based approach at reduced costs and represent a truly disruptive technology that may start a revolution in medicine.
Collapse
|
50
|
Off-on polyadenylation strategy as a supplemental mechanism for silencing toxic transgene expression during lentiviral vector production. Biotechniques 2014; 56:311-2, 314-8. [PMID: 24924391 DOI: 10.2144/000114178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/29/2014] [Indexed: 11/23/2022] Open
Abstract
Many gene therapy strategies rely on lentiviral-mediated transfer and expression of genes coding for toxic proteins. Methods of controlling transgene expression in target cells have been extensively investigated, but comparatively little attention has been given to controlling toxic protein expression in viral vector-producing cells, despite its potential implications for viral production and transduction efficiency. In this work, we tested a new lentiviral vector with a backbone that inhibits transgene mRNA polyadenylation and subsequent transgene expression in vector-producing cells. Transgene mRNA polyadenylation was not affected in transduced cells. In a model using enhanced green fluorescent protein (EGFP) cDNA under the control of the human phosphoglycerate kinase (PGK) promoter, flow cytometry demonstrated that transgene expression was dramatically decreased in 293T cells transfected with this new vector in its plasmid configuration. Viral production was maintained, and expression was fully restored in transduced HuH7 and 293T cells. These results provide the basis for a new strategy to improve the production of lentiviral vectors expressing toxic transgenes.
Collapse
|