85 results on '"Joeri L. Aerts"'
Search Results
2. Multimeric immunotherapeutic complexes activating natural killer cells towards HIV-1 cure
- Author
-
Rafaëla Schober, Bianca Brandus, Thessa Laeremans, Gilles Iserentant, Camille Rolin, Géraldine Dessilly, Jacques Zimmer, Michel Moutschen, Joeri L. Aerts, Xavier Dervillez, and Carole Seguin-Devaux
- Subjects
NK cells ,HIV cure ,Immunotherapy ,IL-15 ,NKG2A ,KIR2DL ,Medicine - Abstract
Abstract Background Combination antiretroviral therapy (cART) has dramatically extended the life expectancy of people living with HIV-1 and improved their quality of life. There is nevertheless no cure for HIV-1 infection since HIV-1 persists in viral reservoirs of latently infected CD4+ T cells. cART does not eradicate HIV-1 reservoirs or restore cytotoxic natural killer (NK) cells which are dramatically reduced by HIV-1 infection, and express the checkpoint inhibitors NKG2A or KIR2DL upregulated after HIV-1 infection. Cytotoxic NK cells expressing the homing receptor CXCR5 were recently described as key subsets controlling viral replication. Methods We designed and evaluated the potency of “Natural killer activating Multimeric immunotherapeutic compleXes”, called as NaMiX, combining multimers of the IL-15/IL-15Rα complex with an anti-NKG2A or an anti-KIR single-chain fragment variable (scFv) to kill HIV-1 infected CD4+ T cells. The oligomerization domain of the C4 binding protein was used to associate the IL-15/IL-15Rα complex to the scFv of each checkpoint inhibitor as well as to multimerize each entity into a heptamer (α form) or a dimer (β form). Each α or β form was compared in different in vitro models using one-way ANOVA and post-hoc Tukey’s tests before evaluation in humanized NSG tg-huIL-15 mice having functional NK cells. Results All NaMiX significantly enhanced the cytolytic activity of NK and CD8+ T cells against Raji tumour cells and HIV-1+ ACH-2 cells by increasing degranulation, release of granzyme B, perforin and IFN-γ. Targeting NKG2A had a stronger effect than targeting KIR2DL due to higher expression of NKG2A on NK cells. In viral inhibition assays, NaMiX initially increased viral replication of CD4+ T cells which was subsequently inhibited by cytotoxic NK cells. Importantly, anti-NKG2A NaMiX enhanced activation, cytotoxicity, IFN-γ production and CXCR5 expression of NK cells from HIV-1 positive individuals. In humanized NSG tg-huIL-15 mice, we confirmed enhanced activation, degranulation, cytotoxicity of NK cells, and killing of HIV-1 infected cells from mice injected with the anti-NKG2A.α NaMiX, as compared to control mice, as well as decreased total HIV-1 DNA in the lung. Conclusions NK cell-mediated killing of HIV-1 infected cells by NaMiX represents a promising approach to support HIV-1 cure strategies.
- Published
- 2023
- Full Text
- View/download PDF
3. Unveiling the intricacies of gene delivery: Caveolae-mediated endocytosis induces efficient mRNA delivery in slow-dividing cells
- Author
-
Sabine den Roover and Joeri L. Aerts
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Published
- 2023
- Full Text
- View/download PDF
4. Autologous dendritic cell vaccination against HIV-1 induces changes in natural killer cell phenotype and functionality
- Author
-
Thessa Laeremans, Sabine den Roover, Cynthia Lungu, Sigrid D’haese, Rob A. Gruters, Sabine D. Allard, and Joeri L. Aerts
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Although natural killer (NK) cells have been studied in connection with dendritic cell (DC)-based vaccination in the field of cancer immunology, their role has barely been addressed in the context of therapeutic vaccination against HIV-1. In this study, we evaluated whether a therapeutic DC-based vaccine consisting of monocyte-derived DCs electroporated with Tat, Rev and Nef encoding mRNA affects NK cell frequency, phenotype and functionality in HIV-1-infected individuals. Although the frequency of total NK cells did not change, we observed a significant increase in cytotoxic NK cells following immunisation. In addition, significant changes in the NK cell phenotype associated with migration and exhaustion were observed together with increased NK cell-mediated killing and (poly)functionality. Our results show that DC-based vaccination has profound effects on NK cells, which highlights the importance of evaluating NK cells in future clinical trials looking at DC-based immunotherapy in the context of HIV-1 infection.
- Published
- 2023
- Full Text
- View/download PDF
5. Unraveling the Effects of a Talimogene Laherparepvec (T-VEC)-Induced Tumor Oncolysate on Myeloid Dendritic Cells
- Author
-
Jens Tijtgat, Jolien De Munck, Inès Dufait, Julia Katharina Schwarze, Ivan Van Riet, Lorenzo Franceschini, Karine Breckpot, Joeri L. Aerts, Bart Neyns, and Sandra Tuyaerts
- Subjects
melanoma ,myeloid dendritic cell ,BDCA-1 ,BDCA-3 ,Talimogene laherparepvec ,cell therapy ,Immunologic diseases. Allergy ,RC581-607 - Abstract
T-VEC, a HSV-1 derived oncolytic virus, is approved for the treatment of advanced melanoma. The mechanisms that underly the systemic anti-tumor effect that is seen following intratumoral injection have not yet been studied but are likely to be mediated by myeloid dendritic cells (myDC) that initiate an adaptive immune response. In this study we could demonstrate that T-VEC is non-toxic for human myDC. T-VEC and a T-VEC oncolysate of melanoma cell lines were able to mature human myDC. myDC were able to take up lysed melanoma cells and cross-present melanoma-derived tumor antigens to antigen-specific T cells. Our results support the possible role of myDC as mediators of an adaptive anti-tumor effect and intratumoral co-administration of T-VEC plus autologous myDC could be a complementary treatment option. A clinical trial that investigates this hypothesis is currently ongoing.
- Published
- 2021
- Full Text
- View/download PDF
6. Intranodal administration of mRNA encoding nucleoprotein provides cross-strain immunity against influenza in mice
- Author
-
Patrick Tjok Joe, Ioanna Christopoulou, Lien van Hoecke, Bert Schepens, Tine Ysenbaert, Carlo Heirman, Kris Thielemans, Xavier Saelens, and Joeri L. Aerts
- Subjects
mRNA ,Influenza ,Universal vaccine ,Intranodal ,Nucleoprotein ,T cell ,Medicine - Abstract
Abstract Background Current human influenza vaccines lack the adaptability to match the mutational rate of the virus and therefore require annual revisions. Because of extensive manufacturing times and the possibility that antigenic alterations occur during viral vaccine strain production, an inherent risk exists for antigenic mismatch between the new influenza vaccine and circulating viruses. Targeting more conserved antigens such as nucleoprotein (NP) could provide a more sustainable vaccination strategy by inducing long term and heterosubtypic protection against influenza. We previously demonstrated that intranodal mRNA injection can induce potent antigen-specific T-cell responses. In this study, we investigated whether intranodal administration of mRNA encoding NP can induce T-cell responses capable of protecting against a heterologous influenza virus challenge. Methods BALB/c mice were immunized in the inguinal lymph nodes with different vaccination regimens of mRNA encoding NP. Immune responses were compared with NP DNA vaccination via IFN-γ ELISPOT and in vivo cytotoxicity. For survival experiments, mice were prime-boost vaccinated with 17 µg NP mRNA and infected with 1LD50 of H1N1 influenza virus 8 weeks after boost. Weight was monitored and viral titers, cytokines and immune cell populations in the bronchoalveolar lavage, and IFN-γ responses in the spleen were analyzed. Results Our results demonstrate that NP mRNA induces superior systemic T-cell responses against NP compared to classical DNA vaccination. These responses were sustained for several weeks even at low vaccine doses. Upon challenge infection, vaccination with NP mRNA resulted in reduced lung viral titers and improved recovery from infection. Finally, we show that vaccination with NP mRNA affects the immune response in infected lungs by lowering immune cell infiltration while increasing the fraction of T cells, monocytes and MHC II+ alveolar macrophages within immune infiltrates. This change was associated with altered levels of both pro- and anti-inflammatory cytokines. Conclusions These findings suggest that intranodal vaccination with NP mRNA induces cross-strain immunity against influenza, but also highlight a paradox of influenza immunity, whereby robust immune responses can provide protection, but can also transiently exacerbate symptoms during infection.
- Published
- 2019
- Full Text
- View/download PDF
7. Efficient Induction of Antigen-Specific CD8+ T-Cell Responses by Cationic Peptide-Based mRNA Nanoparticles
- Author
-
Sigrid D’haese, Thessa Laeremans, Sabine den Roover, Sabine D. Allard, Guido Vanham, and Joeri L. Aerts
- Subjects
mRNA ,nanoparticles ,nanovaccines ,Pharmacy and materia medica ,RS1-441 - Abstract
A major determinant for the success of mRNA-based vaccines is the composition of the nanoparticles (NPs) used for formulation and delivery. Cationic peptides represent interesting candidate carriers for mRNA, since they have been shown to efficiently deliver nucleic acids to eukaryotic cells. mRNA NPs based on arginine-rich peptides have previously been demonstrated to induce potent antigen-specific CD8+ T-cell responses. We therefore compared the histidine-rich amphipathic peptide LAH4-L1 (KKALLAHALHLLALLALHLAHALKKA) to the fully substituted arginine variant (LAH4-L1R) for their capacity to formulate mRNA and transfect dendritic cells (DCs). Although both peptides encapsulated mRNA to the same extent, and showed excellent uptake in DCs, the gene expression level was significantly higher for LAH4-L1. The LAH4-L1–mRNA NPs also resulted in enhanced antigen presentation in the context of MHC I compared to LAH4-L1R in primary murine CD103+ DCs. Both peptides induced DC maturation and inflammasome activation. Subsequent ex vivo stimulation of OT-I splenocytes with transfected CD103+ DCs resulted in a high proportion of polyfunctional CD8+ T cells for both peptides. In addition, in vivo immunization with LAH4-L1 or LAH4-L1R–mRNA NPs resulted in proliferation of antigen-specific T cells. In conclusion, although LAH4-L1 outperformed LAH4-L1R in terms of transfection efficiency, the immune stimulation ex vivo and in vivo was equally efficient.
- Published
- 2022
- Full Text
- View/download PDF
8. Selection of appropriate control genes to assess expression of tumor antigens using real-time RT-PCR
- Author
-
Joeri L. Aerts, Monica I. Gonzales, and Suzanne L. Topalian
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Real-time reverse transcription PCR (RT-PCR) is a sensitive and accurate method to monitor gene expression and is often used to profile the expression of putative tumor antigens in the context of immunotherapy. However, this technique consists of several steps, including cell processing, RNA extraction, RNA storage, assessment of RNA concentration, and cDNA synthesis prior to PCR. To compensate for potential variability introduced in this procedure, the expression of housekeeping genes is commonly assessed in parallel with the expression of the gene of interest. In this study, the expression of a variety of housekeeping genes in a panel of 26 different human tumor and embryonal cell lines was assessed using real-time RT-PCR. For some control genes, the variability in expression was significant between different cell lines, despite the equalization of quantities of input RNA. The greatest variability was found for GAPDH. The lowest variability was found for β-glucuronidase (GUS) and 18S rRNA. While real-time RT-PCR is a powerful tool for gene expression analysis, these results suggest that the choice of control genes to normalize the expression of the gene of interest is critical to the interpretation of experimental results and should be tailored to the nature of the study.
- Published
- 2004
- Full Text
- View/download PDF
9. Evaluation of functional candidate biomarkers of non-genotoxic hepatocarcinogenicity in human liver spheroid co-cultures
- Author
-
Bruna dos Santos Rodrigues, Kaat Leroy, Milos Mihajlovic, Sybren De Boever, Sarah Vanbellingen, Bruno Cogliati, Joeri L. Aerts, Mathieu Vinken, Experimental in vitro toxicology and dermato-cosmetology, Pharmaceutical and Pharmacological Sciences, Faculty of Medicine and Pharmacy, Neuro-Aging & Viro-Immunotherapy, and Teacher Education
- Subjects
Health, Toxicology and Mutagenesis ,General Medicine ,Toxicology - Abstract
Validated in vitro assays for testing non-genotoxic carcinogenic potential of chemicals are currently not available. Consequently, the two-year rodent bioassay remains the gold standard method for the identification of these chemicals. Transcriptomic and proteomic analyses have provided a comprehensive understanding of the non-genotoxic carcinogenic processes, however, functional changes induced by effects at transcriptional and translational levels have not been addressed. The present study was set up to test a number of proposed in vitro biomarkers of non-genotoxic hepatocarcinogenicity at the functional level using a translational 3-dimensional model. Spheroid cultures of human hepatocytes and stellate cells were exposed to 5 genotoxic carcinogenic, 5 non-genotoxic carcinogenic, and 5 non-carcinogenic chemical compounds and assessed for oxidative stress, mitochondrial dysfunction, endoplasmic reticulum stress, apoptosis, and inflammation. The spheroid model could capture many of these events triggered by the genotoxic carcinogenic chemicals, particularly aflatoxin B1 and hydroquinone. Nonetheless, no clear distinction could be made between genotoxic and non-genotoxic hepatocarcinogenicity. Therefore, spheroid cultures of human liver cells may be appropriate in vitro tools for mechanistic investigation of chemical-induced hepatocarcinogenicity, however, these mechanisms and their read-outs do not seem to be eligible biomarkers for detecting non-genotoxic carcinogenic chemicals.
- Published
- 2023
- Full Text
- View/download PDF
10. Qualitative plasma viral load determination as a tool for screening of viral reservoir size in PWH
- Author
-
Thessa Laeremans, Sigrid D’haese, Jonathan Aernout, Kurt Barbé, Pieter Pannus, Sofie Rutsaert, Ellen Vancutsem, Guido Vanham, Coca Necsoi, Ward De Spiegelaere, Marie Couttenye, Natacha Herssens, Marie-Angélique De Scheerder, Stéphane De Wit, Linos Vandekerckhove, Eric Florence, Joeri L. Aerts, Sabine D. Allard, Neuro-Aging & Viro-Immunotherapy, Faculty of Medicine and Pharmacy, Pharmaceutical and Pharmacological Sciences, Radiation Therapy, Artificial Intelligence supported Modelling in clinical Sciences, Biostatistics and medical informatics, Digital Mathematics, Public Health Sciences, Supporting clinical sciences, Microbiology and Infection Control, Clinical Biology, Internal Medicine, and Clinical sciences
- Subjects
Immunology ,HIV Infections/drug therapy ,Health Informatics ,HIV Infections ,Viral Load ,Plasma/chemistry ,Plasma ,Infectious Diseases ,DNA, Viral ,DNA, Viral/analysis ,Internal Medicine ,Leukocytes, Mononuclear ,Humans ,RNA ,RNA, Viral ,Immunology and Allergy ,Human medicine - Abstract
Objective(s): Suppression of viral replication in patients on antiretroviral therapy (ART) is determined by plasma viral load (pVL) measurement. Whenever pVL reaches values below the limit of quantification, the qualitative parameter 'target detected' or 'target not detected' is available but often not reported to the clinician. We investigated whether qualitative pVL measurements can be used to estimate the viral reservoir size. Design: The study recruited 114 people with HIV (PWH) who are stable on ART between 2016 and 2018. The percentage of pVL measurements qualitatively reported as 'target detected' (PTD) within a 2-year period was calculated. Methods: t-DNA and US-RNA were used to estimate viral reservoir size and were quantified on peripheral blood mononuclear cells (PBMCs) using droplet digital PCR. Results: A median of 6.5 pVL measurements over a 2-year period was evaluated for each participant to calculate PTD. A positive correlation was found between t-DNA and PTD (r = 0.24; P = 0.011) but not between US-RNA and PTD (r = 0.1; P = 0.3). A significantly lower PTD was observed in PWH with a small viral reservoir, as estimated by t-DNA less than 66 copies/10(6) PBMCs and US-RNA less than 10 copies/10(6) PBMCs, compared with PWH with a larger viral reservoir (P = 0.001). We also show that t-DNA is detectable whenever PTD is higher than 56% and that ART regimen does not affect PTD. Conclusion: Our study shows that PTD provides an efficient parameter to preselect participants with a small viral reservoir based on already available pVL data for future HIV cure trials.
- Published
- 2022
- Full Text
- View/download PDF
11. Push-Through Filtration of Emulsified Adipose Tissue Over a 500-µm Mesh Significantly Reduces the Amount of Stromal Vascular Fraction and Mesenchymal Stem Cells
- Author
-
Lisa Ramaut, Linde Moonen, Thessa Laeremans, Joeri L Aerts, Maxim Geeroms, and Moustapha Hamdi
- Subjects
Surgery ,General Medicine - Abstract
Background Mechanical isolation of stromal vascular fraction (SVF) intends to isolate the stromal component from the parenchymal cells. Emulsification is currently the most common used disaggregation method and is effective in disrupting adipocytes and fragmenting extracellular matrix. Subsequent push-through filtration of emulsified adipose tissue removes parts of the extracellular matrix (ECM) that is not sufficiently micronized to further liquify the tissue. Objectives Filtration over a 500 µm mesh filter might affect the SVF and adipose derived mesenchymal stem cell (MSC) quantity in emulsified lipoaspirates samples by removing ECM fragments. Methods Eleven lipoaspirate samples from healthy non-obese women were harvested and emulsified in thirty passes. One half of the sample was filtered through a 500 µm mesh filter while the other half was left unfiltered. Paired samples were processed and analyzed by flowcytometry to identify cellular viability, SVF and MSC yield. Results Push-through filtration reduced the amount of SVF cells by on average 39.65 ± 5,67% (p Conclusions This study has shown that retention of fibrous remnants by push through filters removes ECM containing SVF and MSC from emulsified lipoaspirates. Processing methods should aim either to further micronize the lipoaspirate before filtering or to not filter the samples at all, to preserve both the cellular component carried within the ECM as the inductive properties of ECM itself.
- Published
- 2023
- Full Text
- View/download PDF
12. Lifespan extension with preservation of hippocampal function in aged system xc−-deficient male mice
- Author
-
Lise Verbruggen, Gamze Ates, Olaya Lara, Jolien De Munck, Agnès Villers, Laura De Pauw, Sigrid Ottestad-Hansen, Sho Kobayashi, Pauline Beckers, Pauline Janssen, Hideyo Sato, Yun Zhou, Emmanuel Hermans, Rose Njemini, Lutgarde Arckens, Niels C. Danbolt, Dimitri De Bundel, Joeri L. Aerts, Kurt Barbé, Benoit Guillaume, Laurence Ris, Eduard Bentea, Ann Massie, Faculty of Medicine and Pharmacy, Pharmaceutical and Pharmacological Sciences, Neuro-Aging & Viro-Immunotherapy, Gerontology, Frailty in Ageing, Experimental Pharmacology, Radiation Therapy, Artificial Intelligence supported Modelling in clinical Sciences, Biostatistics and medical informatics, Digital Mathematics, Public Health Sciences, and UCL - SSS/IONS/CEMO - Pôle Cellulaire et moléculaire
- Subjects
male mice ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Neuroscience(all) ,Lifespan extension ,Pharmaceutical Science ,Health Informatics ,Geriatrics and Gerontology ,preservation of hippocampal function ,Molecular Biology ,aged system x c--deficient - Abstract
The cystine/glutamate antiporter system xc− has been identified as the major source of extracellular glutamate in several brain regions as well as a modulator of neuroinflammation, and genetic deletion of its specific subunit xCT (xCT−/−) is protective in mouse models for age-related neurological disorders. However, the previously observed oxidative shift in the plasma cystine/cysteine ratio of adult xCT−/− mice led to the hypothesis that system xc− deletion would negatively affect life- and healthspan. Still, till now the role of system xc− in physiological aging remains unexplored. We therefore studied the effect of xCT deletion on the aging process of mice, with a particular focus on the immune system, hippocampal function, and cognitive aging. We observed that male xCT−/− mice have an extended lifespan, despite an even more increased plasma cystine/cysteine ratio in aged compared to adult mice. This oxidative shift does not negatively impact the general health status of the mice. On the contrary, the age-related priming of the innate immune system, that manifested as increased LPS-induced cytokine levels and hypothermia in xCT+/+ mice, was attenuated in xCT−/− mice. While this was associated with only a very moderate shift towards a more anti-inflammatory state of the aged hippocampus, we observed changes in the hippocampal metabolome that were associated with a preserved hippocampal function and the retention of hippocampus-dependent memory in male aged xCT−/− mice. Targeting system xc− is thus not only a promising strategy to prevent cognitive decline, but also to promote healthy aging.
- Published
- 2022
- Full Text
- View/download PDF
13. Supplementary Figure Legends 1-2 from Proinflammatory Characteristics of SMAC/DIABLO-Induced Cell Death in Antitumor Therapy
- Author
-
Karine Breckpot, Joeri L. Aerts, Kris Thielemans, Carlo Heirman, Tomas Bos, Iain A. McNeish, Anje Cauwels, Sarah Maenhout, Cleo Goyvaerts, Sandra Van Lint, and Perpetua U. Emeagi
- Abstract
PDF file - 60K
- Published
- 2023
- Full Text
- View/download PDF
14. Data from Quantifying the Activity of Adenoviral E1A CR2 Deletion Mutants Using Renilla Luciferase Bioluminescence and 3′-Deoxy-3′-[18F]Fluorothymidine Positron Emission Tomography Imaging
- Author
-
Iain A. McNeish, Nicholas R. Lemoine, Eric O. Aboagye, Sarah K. Baird, Joeri L. Aerts, Michelle Lockley, and Julius Leyton
- Abstract
The adenoviral E1A CR2 mutant dl922-947 has potent activity in ovarian cancer. We have used Renilla luciferase bioluminescence imaging to monitor viral E1A expression and replication and [18F]fluorothymidine positron emission tomography ([18F]FLT-PET) to quantify the activity of dl922-947 in vivo. We created dlCR2 Ren, with the same E1A CR2 deletion as dl922-947 and the luciferase gene from Renilla reniformis downstream of E1. Light emitted from s.c. and i.p. IGROV1 ovarian carcinoma xenografts was measured following treatment with dlCR2 Ren. Mice bearing s.c. IGROV1 xenografts were injected with 2.96 to 3.7 MBq of [18F]FLT 48 and 168 hours following i.t. injection of dl922-947 or control virus Ad LM-X. The presence of Renilla luciferase in dlCR2 Ren did not reduce in vitro nor in vivo potency compared with dl922-947. Light emission correlated closely with E1A expression in vitro and peaked 48 hours after dlCR2 Ren injection in both s.c. and i.p. IGROV1 xenografts. It diminished by 168 hours in s.c. tumors but persisted for at least 2 weeks in i.p. models. Normalized tumor [18F]FLT uptake at 60 minutes (NUV60), fractional retention, and area under radioactivity curve all decreased marginally 48 hours after dl922-947 treatment and significantly at 168 hours compared with controls. There was a close linear correlation between NUV60 and both tumor proliferation (Ki67 labeling index) and thymidine kinase 1 expression. Renilla luciferase bioluminescence and [18F]FLT-PET imaging are capable of quantifying the activity and effectiveness of E1A CR2–deleted adenoviral mutants in ovarian cancer. (Cancer Res 2006; 66(18): 9178-85)
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure 2 from Proinflammatory Characteristics of SMAC/DIABLO-Induced Cell Death in Antitumor Therapy
- Author
-
Karine Breckpot, Joeri L. Aerts, Kris Thielemans, Carlo Heirman, Tomas Bos, Iain A. McNeish, Anje Cauwels, Sarah Maenhout, Cleo Goyvaerts, Sandra Van Lint, and Perpetua U. Emeagi
- Abstract
PDF file - 117K, Uptake of LV-tSMAC transduced dying human melanoma cells enables dendritic cells to activate tumor-infiltrating lymphocytes.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Figures 1-3 from Quantifying the Activity of Adenoviral E1A CR2 Deletion Mutants Using Renilla Luciferase Bioluminescence and 3′-Deoxy-3′-[18F]Fluorothymidine Positron Emission Tomography Imaging
- Author
-
Iain A. McNeish, Nicholas R. Lemoine, Eric O. Aboagye, Sarah K. Baird, Joeri L. Aerts, Michelle Lockley, and Julius Leyton
- Abstract
Supplementary Figures 1-3 from Quantifying the Activity of Adenoviral E1A CR2 Deletion Mutants Using Renilla Luciferase Bioluminescence and 3′-Deoxy-3′-[18F]Fluorothymidine Positron Emission Tomography Imaging
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figure 1 from Proinflammatory Characteristics of SMAC/DIABLO-Induced Cell Death in Antitumor Therapy
- Author
-
Karine Breckpot, Joeri L. Aerts, Kris Thielemans, Carlo Heirman, Tomas Bos, Iain A. McNeish, Anje Cauwels, Sarah Maenhout, Cleo Goyvaerts, Sandra Van Lint, and Perpetua U. Emeagi
- Abstract
PDF file - 184K, The therapeutic effect of LV-tSMAC involves effector T-cells.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Methods from Quantifying the Activity of Adenoviral E1A CR2 Deletion Mutants Using Renilla Luciferase Bioluminescence and 3′-Deoxy-3′-[18F]Fluorothymidine Positron Emission Tomography Imaging
- Author
-
Iain A. McNeish, Nicholas R. Lemoine, Eric O. Aboagye, Sarah K. Baird, Joeri L. Aerts, Michelle Lockley, and Julius Leyton
- Abstract
Supplementary Methods from Quantifying the Activity of Adenoviral E1A CR2 Deletion Mutants Using Renilla Luciferase Bioluminescence and 3′-Deoxy-3′-[18F]Fluorothymidine Positron Emission Tomography Imaging
- Published
- 2023
- Full Text
- View/download PDF
19. Dendritic cell immunotherapy followed by cART interruption during HIV-1 infection induces plasma protein markers of cellular immunity and neutrophil recruitment.
- Author
-
Henk-Jan van den Ham, Jason D Cooper, Jakub Tomasik, Sabine Bahn, Joeri L Aerts, Albert D M E Osterhaus, Rob A Gruters, Arno C Andeweg, and DC-TRN trial investigators
- Subjects
Medicine ,Science - Abstract
To characterize the host response to dendritic cell-based immunotherapy and subsequent combined antiretroviral therapy (cART) interruption in HIV-1-infected individuals at the plasma protein level.An autologous dendritic cell (DC) therapeutic vaccine was administered to HIV-infected individuals, stable on cART. The effect of vaccination was evaluated at the plasma protein level during the period preceding cART interruption, during analytical therapy interruption and at viral reactivation. Healthy controls and post-exposure prophylactically treated healthy individuals were included as controls.Plasma marker ('analyte') levels including cytokines, chemokines, growth factors, and hormones were measured in trial participants and control plasma samples using a multiplex immunoassay. Analyte levels were analysed using principle component analysis, cluster analysis and limma. Blood neutrophil counts were analysed using linear regression.Plasma analyte levels of HIV-infected individuals are markedly different from those of healthy controls and HIV-negative individuals receiving post-exposure prophylaxis. Viral reactivation following cART interruption also affects multiple analytes, but cART interruption itself only has only a minor effect. We find that Thyroxine-Binding Globulin (TBG) levels and late-stage neutrophil numbers correlate with the time off cART after DC vaccination. Furthermore, analysis shows that cART alters several regulators of blood glucose levels, including C-peptide, chromogranin-A and leptin. HIV reactivation is associated with the upregulation of CXCR3 ligands.Chronic HIV infection leads to a change in multiple plasma analyte levels, as does virus reactivation after cART interruption. Furthermore, we find evidence for the involvement of TBG and neutrophils in the response to DC-vaccination in the setting of HIV-infection.
- Published
- 2018
- Full Text
- View/download PDF
20. Natural Killer activating multimeric immunotherapeutic complexes (NaMiX) induce cytotoxic activity and killing of HIV-1 infected cells
- Author
-
Rafaela Schober, Bianca Brandus, Thessa Laeremans, Gilles Iserentant, Géraldine Dessilly, Jacques Zimmer, Michel Moutschen, Joeri L Aerts, Xavier Dervillez, and Carole Seguin-Devaux
- Abstract
HIV-1 persists in viral reservoirs of latently infected CD4+T cells containing integrated replication-competent viral DNA. Combined Antiretroviral Therapy (cART) does not eradicate HIV-1 reservoirs and treatment interruption will ultimately lead to viral load rebound. HIV-1 infection dramatically reduces the proportion of functional NK cell subsets and increases the expression of the checkpoint inhibitors NKG2A and KIR2DL. In this regard, we developed novel recombinant molecules combining multimers of the IL-15/IL-15Rα complex with the single-chain fragment variables (scFvs) of NKG2A or KIR2DL, and named them as Natural killer activating Multimeric immunotherapeutic compleXes (NaMiX). NaMiX significantly improved the cytotoxic activity of NK cells against HIV-1 positive ACH-2 cells and resistant Raji cancer cells by increasing their degranulation capacity, release of granzyme B, perforin and IFN-γ expression. Targeting the NKG2A receptor had a stronger effect compared to the targeting of the KIR2DL receptor due to its higher expression on NK cells. In a viral inhibition assay using CD4+T cells from HIV-1 positive patients under cART, NaMiX initially increased viral replication which was subsequently inhibited by stimulated NK cells. In humanized NSG tg-huIL-15 mice showing functional NK cells, we observed enhanced activation, degranulation and killing by NK cells from the spleen of mice treated with anti-NKG2A NaMiX compared to the cells of control mice previously infected with HIV-1 and treated with cART. Although NaMiX did not delay viral load rebound after treatment interruption in a first attempt, it tend to decrease total HIV-1 DNA in the lungs of the mice. Blocking the inhibitory receptor NKG2A in combination with targeted multimers of IL-15 on NK cells could therefore be a promising immunotherapeutic strategy towards HIV-1 functional cure.
- Published
- 2022
- Full Text
- View/download PDF
21. Adjuvants, the Elephant in the Room for RNA Vaccines
- Author
-
Sigrid D’haese, Sabine den Roover, Joeri L. Aerts, Neuro-Aging & Viro-Immunotherapy, Pharmaceutical and Pharmacological Sciences, and Faculty of Medicine and Pharmacy
- Abstract
Adjuvants are crucial components of vaccines. Nevertheless, they are frequently considered as mere “excipients”, and their mode of action is often poorly understood. Although the attractiveness of mRNA as an immunogen has been recognized already more than thirty years ago, it wasn’t until the current COVID-19 crisis that its full potential was shown. From a fringe approach, it has now become a leading technology in vaccine development which will no doubt result in a tremendous boost in both prophylactic and therapeutic vaccination settings. The issue of finding the right adjuvant is especially relevant for mRNA-based vaccines, as mRNA itself is a strong activator of innate immune responses which represents a doubleedged sword. Moreover, given the high sensitivity of RNA to ambient RNases, and to improve delivery efficiency, in recent years, a lot of effort has been invested in developing ways to package the mRNA in so-called nanoparticle formulations. Currently approved mRNA-based vaccines are all formulatedin lipid nanoparticles, but many other approaches are being explored, each of which will result in a different type of immune stimulation. In this chapter, we want to provide an overview of the potential adjuvant effect of different types of nanoparticles and implications for vaccine development.
- Published
- 2022
- Full Text
- View/download PDF
22. Oncolytic Herpes Simplex Virus Type 1 Induces Immunogenic Cell Death Resulting in Maturation of BDCA-1+ Myeloid Dendritic Cells
- Author
-
Philipp Kalus, Jolien De Munck, Sarah Vanbellingen, Laura Carreer, Thessa Laeremans, Katrijn Broos, Inès Dufait, Julia K. Schwarze, Ivan Van Riet, Bart Neyns, Karine Breckpot, Joeri L. Aerts, Neuro-Aging & Viro-Immunotherapy, Faculty of Medicine and Pharmacy, Pharmaceutical and Pharmacological Sciences, Teacher Education, Clinical sciences, Radiation Therapy, Laboratory of Molecular and Medical Oncology, Internal Medicine, Basic (bio-) Medical Sciences, Hematology, Medical Oncology, and Laboratory of Molecullar and Cellular Therapy
- Subjects
Oncolytic Virotherapy ,Medicine(all) ,cancer immunotherapy ,Organic Chemistry ,General Medicine ,Myeloid dendritic cells ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,immunogenic cell death ,melanoma ,Physical and Theoretical Chemistry ,Molecular Biology ,oncolytic virus (OV) ,immunogenic cell death (ICD) ,myeloid DCs (myDCs) ,Spectroscopy - Abstract
Recently, a paradigm shift has been established for oncolytic viruses (OVs) as it was shown that the immune system plays an important role in the specific killing of tumor cells by OVs. OVs have the intrinsic capacity to provide the right signals to trigger anti-tumor immune responses, on the one hand by delivering virus-derived innate signals and on the other hand by inducing immunogenic cell death (ICD), which is accompanied by the release of various damage-associated molecules from infected tumor cells. Here, we determined the ICD-inducing capacity of Talimogene laherparepvec (T-VEC), a herpes simplex virus type 1 based OV, and benchmarked this to other previously described ICD (e.g., doxorubicin) and non-ICD inducing agents (cisplatin). Furthermore, we studied the capability of T-VEC to induce the maturation of human BDCA-1+ myeloid dendritic cells (myDCs). We found that T-VEC treatment exerts direct and indirect anti-tumor effects as it induces tumor cell death that coincides with the release of hallmark mediators of ICD, while simultaneously contributing to the maturation of BDCA-1+ myDCs. These results unequivocally cement OVs in the category of cancer immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
23. T-cell subsets in the skin and their role in inflammatory skin disorders
- Author
-
Inge Kortekaas Krohn, Femke van Wijk, Jan Gutermuth, Cleo Goyvaerts, Karine Breckpot, Edward F. Knol, Joeri L. Aerts, Dermatology, Neuro-Aging & Viro-Immunotherapy, Pharmaceutical and Pharmacological Sciences, Basic (bio-) Medical Sciences, Laboratory of Molecullar and Cellular Therapy, Artificial Intelligence supported Modelling in clinical Sciences, Gerontology, and Skin function and permeability
- Subjects
skin ,medicine.medical_treatment ,T cell ,Immunology ,Skin Diseases ,DISEASE ,Allergic inflammation ,Chemokine receptor ,T-Lymphocyte Subsets ,melanoma ,Humans ,Immunology and Allergy ,Medicine ,Receptor ,Transcription factor ,Inflammation ,business.industry ,Acquired immune system ,Cytokine ,medicine.anatomical_structure ,Health ,Cytokines ,business ,Function (biology) - Abstract
T lymphocytes (T cells) are major players of the adaptive immune response. Naive T cells are primed in the presence of cytokines, leading to polarization into distinct T-cell subsets with specific functions. These subsets are classified based on their T-cell receptor profile, expression of transcription factors, surface cytokine and chemokine receptors, and their cytokine production, which together determine their specific function. This review provides an overview of the various T-cell subsets and their function in several inflammatory skin disorders ranging from allergic inflammation to skin tumors. Moreover, we highlight similarities of T-cell responses across different skin disorders, demonstrating the presence of similar and opposing functions for the different T-cell subsets. Finally, we discuss the effects of currently available and promising therapeutic approaches to harness T cells in inflammatory skin diseases for which efficacy next to unwanted side effects provide new insights into the pathophysiology of skin disorders.
- Published
- 2022
24. Layer-by-Layer technique as a versatile tool for gene delivery applications
- Author
-
Joeri L. Aerts, Dmitrii S. Linnik, Alexander S. Timin, Mikhail V. Zyuzin, Gleb B. Sukhorukov, Kirill V. Lepik, Yana V. Tarakanchikova, Neuro-Aging & Viro-Immunotherapy, Pharmaceutical and Pharmacological Sciences, and Laboratory of Molecullar and Cellular Therapy
- Subjects
templates ,Computer science ,Genetic enhancement ,Genetic Vectors ,clinical perspective ,Pharmaceutical Science ,Nanotechnology ,Layer-by-Layer ,02 engineering and technology ,Gene delivery ,ex vivo cell modification ,030226 pharmacology & pharmacy ,03 medical and health sciences ,Broad spectrum ,Pharmacology, Toxicology and Pharmaceutics(all) ,0302 clinical medicine ,Immunology and Microbiology(all) ,RNA, Messenger ,gene delivery ,multilayer capsules ,Gene Editing ,Biochemistry, Genetics and Molecular Biology(all) ,Layer by layer ,Gene Transfer Techniques ,Genetic Therapy ,DNA ,021001 nanoscience & nanotechnology ,RNA ,0210 nano-technology - Abstract
Introduction: Gene therapy is a breakthrough medical field which focuses on the therapeutic delivery of recombinant nucleic acids in order to treat or prevent a broad spectrum of diseases. However, a number of important obstacles remain before its wide introduction into clinical practice can be envisaged. One of the biggest bottlenecks is the lack of efficient and safe delivery technologies, particularly, for in vivo distribution. Above and beyond standard requirements for carriers, the delivery systems for gene therapy ideally use a hit-and-run principle (to minimize off-target effect and display of immunogenic moieties). None of the currently used viral vectors fulfills all of these requirements. Therefore, the growing variety of non-viral delivery platforms represents a promising alternative. Areas covered: This review summarizes the Layer-by-Layer (LbL) approaches that can be effectively used for the gene delivery, considering various examples with the transfer of pDNA, mRNA, siRNA as well as genome-editing tools. Ex vivo gene modification of clinically relevant cells and clinical aspects for possible application of LbL systems in gene therapy are also underlined. Expert opinion: The LbL technique provides broad opportunities for the delivery of genetic material for various purposes and offers promise for future clinical application in gene therapy.
- Published
- 2021
- Full Text
- View/download PDF
25. Lifespan extension with preservation of hippocampal function in aged system x
- Author
-
Lise, Verbruggen, Gamze, Ates, Olaya, Lara, Jolien, De Munck, Agnès, Villers, Laura, De Pauw, Sigrid, Ottestad-Hansen, Sho, Kobayashi, Pauline, Beckers, Pauline, Janssen, Hideyo, Sato, Yun, Zhou, Emmanuel, Hermans, Rose, Njemini, Lutgarde, Arckens, Niels C, Danbolt, Dimitri, De Bundel, Joeri L, Aerts, Kurt, Barbé, Benoit, Guillaume, Laurence, Ris, Eduard, Bentea, and Ann, Massie
- Subjects
Male ,Mice, Inbred C57BL ,Mice ,Amino Acid Transport System y+ ,Longevity ,Animals ,Cystine ,Glutamic Acid ,Cysteine ,Hippocampus - Abstract
The cystine/glutamate antiporter system x
- Published
- 2021
26. Unraveling the Effects of a Talimogene Laherparepvec (T-VEC)-Induced Tumor Oncolysate on Myeloid Dendritic Cells
- Author
-
Inès Dufait, Ivan Van Riet, Joeri L. Aerts, Jens Tijtgat, Sandra Tuyaerts, Julia Katharina Schwarze, Karine Breckpot, Jolien De Munck, Lorenzo Franceschini, Bart Neyns, Laboratory of Molecular and Medical Oncology, Medical Oncology, Clinical sciences, Faculty of Medicine and Pharmacy, Neuro-Aging & Viro-Immunotherapy, Pharmaceutical and Pharmacological Sciences, Radiation Therapy, Internal Medicine, Basic (bio-) Medical Sciences, Hematology, and Laboratory of Molecullar and Cellular Therapy
- Subjects
Myeloid ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Herpesvirus 1, Human ,Myeloid dendritic cells ,Adaptive Immunity ,Lymphocyte Activation ,Antigens, CD1 ,Antineoplastic Agents, Immunological ,Cross-Priming ,Antigen ,Phagocytosis ,Antigens, Neoplasm ,medicine ,melanoma ,Immunology and Allergy ,Humans ,Myeloid Cells ,BDCA-1 ,BDCA-3 ,Talimogene laherparepvec ,Glycoproteins ,Original Research ,Oncolytic Virotherapy ,Biological Products ,business.industry ,hematology ,Melanoma ,Immunotherapy ,Dendritic Cells ,RC581-607 ,myeloid dendritic cell ,medicine.disease ,Acquired immune system ,Oncolytic virus ,medicine.anatomical_structure ,oncology ,Cancer research ,immunotherapy ,Immunologic diseases. Allergy ,cell therapy ,business ,Oncolysate - Abstract
T-VEC, a HSV-1 derived oncolytic virus, is approved for the treatment of advanced melanoma. The mechanisms that underly the systemic anti-tumor effect that is seen following intratumoral injection have not yet been studied but are likely to be mediated by myeloid dendritic cells (myDC) that initiate an adaptive immune response. In this study we could demonstrate that T-VEC is non-toxic for human myDC. T-VEC and a T-VEC oncolysate of melanoma cell lines were able to mature human myDC. myDC were able to take up lysed melanoma cells and cross-present melanoma-derived tumor antigens to antigen-specific T cells. Our results support the possible role of myDC as mediators of an adaptive anti-tumor effect and intratumoral co-administration of T-VEC plus autologous myDC could be a complementary treatment option. A clinical trial that investigates this hypothesis is currently ongoing. ispartof: FRONTIERS IN IMMUNOLOGY vol:12 ispartof: location:Switzerland status: published
- Published
- 2021
27. Off the beaten path
- Author
-
Montserrat Plana, Joeri L. Aerts, Sigrid D'haese, Bernard Verrier, Felipe García, Céline Lacroix, Simona Ruta, Guido Vanham, Pharmaceutical and Pharmacological Sciences, Faculty of Medicine and Pharmacy, Pathologic Biochemistry and Physiology, Laboratory of Molecullar and Cellular Therapy, Vrije Universiteit Brussel [Bruxelles] (VUB), Laboratoire de Biologie Tissulaire et d'ingénierie Thérapeutique UMR 5305 (LBTI), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona (UB), University of Medicine and Pharmacy 'Carol Davila' Bucharest (UMPCD), and Institute of Tropical Medicine [Antwerp] (ITM)
- Subjects
medicine.medical_treatment ,Neuroscience(all) ,Pharmaceutical Science ,HIV Infections ,02 engineering and technology ,Cancer Vaccines ,Epitope ,03 medical and health sciences ,Immune system ,Antigen ,[SDV.MHEP.MI]Life Sciences [q-bio]/Human health and pathology/Infectious diseases ,Virology ,Medicine ,Humans ,RNA, Messenger ,[SDV.IB.BIO]Life Sciences [q-bio]/Bioengineering/Biomaterials ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,0303 health sciences ,business.industry ,Pharmacology. Therapy ,Vaccination ,therapeutic vaccination ,mRNA vaccination ,HIV ,[SDV.MHEP.HEG]Life Sciences [q-bio]/Human health and pathology/Hépatology and Gastroenterology ,Immunotherapy ,[SDV.IMM.IMM]Life Sciences [q-bio]/Immunology/Immunotherapy ,Dendritic Cells ,021001 nanoscience & nanotechnology ,Chemistry ,[CHIM.POLY]Chemical Sciences/Polymers ,[SDV.SP.PG]Life Sciences [q-bio]/Pharmaceutical sciences/Galenic pharmacology ,Immunization ,Immunology ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,nanoparticles ,[SDV.IMM.VAC]Life Sciences [q-bio]/Immunology/Vaccinology ,0210 nano-technology ,business ,Adjuvant ,Ex vivo - Abstract
Over the last few years, immunotherapy for HIV in general and therapeutic vaccination in particular, has received a tremendous boost, both in preclinical research and in clinical applications. This interest is based on the evidence that the immune system plays a crucial role in controlling HIV infection, as shown for long-term non-progressors and elite controllers, and that immune responses can be manipulated towards targeting conserved epitopes. So far, the most successful approach has been vaccination with autologous dendritic cells (DCs) loaded ex vivo with antigens and activation signals. Although this approach offers much promise, it also comes with significant drawbacks such as the requirement of a specialized infrastructure and expertise, as well as major challenges for logistics and storage, making it extremely time consuming and costly. Therefore, methods are being developed to avoid the use of ex vivo generated, autologous DCs. One of these methods is based on mRNA for therapeutic vaccination. mRNA has proven to be a very promising vaccine platform, as the coding information for any desired protein, including antigens and activation signals, can be generated in a very short period of time, showing promise both as an off-the-shelf therapy and as a personalized approach. However, an important drawback of this approach is the short half-life of native mRNA, due to the presence of ambient RNases. In addition, proper immunization requires that the antigens are expressed, processed and presented at the right immunological site (e.g. the lymphoid tissues). An ambivalent aspect of mRNA as a vaccine is its capacity to induce type I interferons, which can have beneficial adjuvant effects, but also deleterious effects on mRNA stability and translation. Thus, proper formulation of the mRNA is crucially important. Many approaches for RNA formulation have already been tested, with mixed success. In this review we discuss the state-of-the-art and future trends for mRNA-nanoparticle formulations for HIV vaccination, both in the prophylactic and in the therapeutic setting.
- Published
- 2021
- Full Text
- View/download PDF
28. Neuroprotection by Insulin-like Growth Factor-1 in Rats with Ischemic Stroke is Associated with Microglial Changes and a Reduction in Neuroinflammation
- Author
-
Erik Boddeke, Joeri L. Aerts, Ahmad Serhan, Ron Kooijman, Faculty of Medicine and Pharmacy, Experimental Pharmacology, Pharmaceutical and Pharmacological Sciences, Laboratory of Molecullar and Cellular Therapy, Basic (bio-) Medical Sciences, Translational Immunology Groningen (TRIGR), and Molecular Neuroscience and Ageing Research (MOLAR)
- Subjects
Male ,INVASION ,Anti-Inflammatory Agents ,Nitric Oxide Synthase Type II ,microglia ,Brain damage ,Pharmacology ,Neuroprotection ,Brain Ischemia ,BRAIN-DAMAGE ,ALTEPLASE ,medicine ,ischemic stroke ,Animals ,Insulin-Like Growth Factor I ,Rats, Wistar ,Stroke ,Neuroinflammation ,NEUTROPHILS ,Medicine(all) ,Microglia ,biology ,RECEPTOR ,business.industry ,ACTIVATED MICROGLIA ,General Neuroscience ,Interleukin ,Brain ,Infarction, Middle Cerebral Artery ,Minocycline ,IMPAIRMENT ,medicine.disease ,IGF-I ,Nitric oxide synthase ,Disease Models, Animal ,medicine.anatomical_structure ,Neuroprotective Agents ,FOCAL CEREBRAL-ISCHEMIA ,inflammation ,biology.protein ,IGF-1 ,neuroprotection ,medicine.symptom ,business ,medicine.drug - Abstract
We and others have shown that insulin-like growth factor-1 (IGF-1) is neuroprotective when administered systemically shortly following stroke. In the current study, we addressed the hypothesis that microglia mediate neuroprotection by IGF-1 following ischemic stroke. Furthermore, we investigated whether IGF-1 modulates pro- and anti-inflammatory mediators in ischemic brain with a special reference to microglia. Ischemic stroke was induced in normal conscious Wistar rats by infusing the vasoconstrictor, endothelin-1 (Et-1), next to middle cerebral artery (MCA). IGF-1 (300 μg) was injected subcutaneously (SC) at 30 and 120 min following stroke. Microglial inhibitor, minocycline, was injected intraperitoneally (IP) at 1 h before stroke (25 mg/kg) and 11 h after stroke (45 mg/kg). Post-stroke IGF-1 treatment reduced the infarct size and increased the sensorimotor function which coincided with an increase in the number of ameboid microglia in the ischemic cortex. Minocycline treatment abrogated the increase in ameboid microglia by IGF-1, while the effect of IGF-1 in the reduction of infarct size was only partially affected. IGF-1 suppressed mRNA expression of inducible nitric oxide synthase (iNOS) and interleukin (IL)-1β in the ischemic hemisphere, while in purified microglia, only iNOS expression levels were reduced. Our findings show that microglia are a target for IGF-1 and that neuroprotection by IGF-1 coincides with down-regulation of inflammatory mediators which could be instrumental to the beneficial effects.
- Published
- 2020
- Full Text
- View/download PDF
29. Rapid viral rebound after analytical treatment interruption in patients with very small HIV reservoir and minimal on-going viral transcription
- Author
-
Ward De Spiegelaere, Joeri L. Aerts, Guido Vanham, Natacha Herssens, Coca Nescoi, Basiel Cole, Linos Vandekerckhove, Eric Florence, Marie-Angélique De Scheerder, Marie-Madeleine Couttenye, Sofie Rutsaert, Sabine Allard, Stéphane De Wit, Pieter Pannus, Achilleas Tsoumanis, Medical Genetics, Clinical sciences, Internal Medicine, Pathologic Biochemistry and Physiology, Pharmaceutical and Pharmacological Sciences, and Laboratory of Molecullar and Cellular Therapy
- Subjects
CD4-Positive T-Lymphocytes ,Male ,Viral rebound ,viral reservoir ,Transcription, Genetic ,Human immunodeficiency virus (HIV) ,HIV Infections ,Virus Replication ,medicine.disease_cause ,0302 clinical medicine ,ANTIRETROVIRAL THERAPY ,Transcription (biology) ,Medicine and Health Sciences ,Medicine ,030212 general & internal medicine ,Research Articles ,treatment interruption ,Remission Induction ,Middle Aged ,Viral Load ,TIME ,PREDICTS ,Infectious Diseases ,PCR ,Female ,post-treatment control ,0305 other medical science ,Viral load ,Research Article ,Adult ,medicine.medical_specialty ,MAINTAINS ,Peripheral blood mononuclear cell ,DNA LEVEL ,03 medical and health sciences ,Internal medicine ,Humans ,Adverse effect ,cell-associated HIV RNA ,030505 public health ,business.industry ,Public Health, Environmental and Occupational Health ,post‐treatment control ,cell‐associated HIV RNA ,RNA ,HIV ,Treatment interruption ,DNA, Viral ,CELLS ,HIV-1 ,Leukocytes, Mononuclear ,Human medicine ,business ,total HIV DNA - Abstract
Introduction Viral remission after analytical treatment interruption (ATI), termed post-treatment control, has been described in a small proportion of HIV-positive patients. This phenomenon has been separately associated to both low levels of HIV-1 proviral DNA as well as cell-associated RNA. We investigated whether the combination of both parameters could help predict delayed viral rebound after treatment interruption (TI). Methods We conducted an open single-arm ATI study in four Belgian HIV reference centres from January 2016 to July 2018. Eligible participants were adults who had fewer than 50 HIV-1 RNA copies/mL for more than two years, more than 500 CD4 cells/mu L for more than three months, and were in general good health. Consenting participants who had fewer than 66 copies total HIV-1 DNA (t-DNA) and fewer than 10 copies cell-associated HIV-1 unspliced RNA (US-RNA) per million peripheral blood mononuclear cells (PBMCs), interrupted therapy and were monitored closely. Antiretroviral therapy (ART) was resumed after two consecutive viral loads exceeding 1000 copies or one exceeding 10,000 copies/mL. The primary outcome was the proportion of participants with fewer than 50 HIV-1 RNA copies/mL 48 weeks after TI. Secondary outcomes were time to viral rebound, the frequency of serious adverse events (AEs) and evolution of t-DNA and US-RNA after TI. Results All 16 consenting participants who interrupted therapy experienced rapid viral rebound two to eight weeks after TI. No serious AEs were observed. Levels of t-DNA and US-RNA increased after TI but returned to pre-ATI levels after treatment restart. None of the studied demographic, clinical and biological parameters were predictive of time of viral rebound. Conclusions The combination of low levels of t-DNA and US-RNA in PBMCs, corresponding respectively to a small and transcriptionally silent viral reservoir, is not predictive of viral remission after TI in patients on ART.
- Published
- 2020
- Full Text
- View/download PDF
30. Potential of memory T cells in bridging preoperative chemoradiation and immunotherapy in rectal cancer
- Author
-
Mark De Ridder, Joeri L. Aerts, Olivier Feron, Sven de Mey, Inès Dufait, Heng Jiang, Hui Wang, Valeri N. Verovski, Thierry Gevaert, Benedikt Engels, UCL - SSS/IREC - Institut de recherche expérimentale et clinique, and UCL - SSS/IREC/FATH - Pôle de Pharmacologie et thérapeutique
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Colorectal cancer ,medicine.medical_treatment ,Antineoplastic Agents ,Disease ,CD8-Positive T-Lymphocytes ,Memory T cells ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Internal medicine ,Preoperative Care ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Rectal cancer ,Neoplasm Staging ,Rectal Neoplasms ,business.industry ,TOR Serine-Threonine Kinases ,IL-2 ,Chemoradiotherapy ,Off-Label Use ,Hematology ,Immunotherapy ,medicine.disease ,Warburg effect ,Total mesorectal excision ,Neoadjuvant Therapy ,Metformin ,Treatment Outcome ,030104 developmental biology ,Clinical research ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,mTOR ,Tumor Escape ,Radiotherapy, Intensity-Modulated ,business ,Adjuvant - Abstract
The management of locally advanced rectal cancer has passed a long way of developments, where total mesorectal excision and preoperative radiotherapy are crucial to secure clinical outcome. These and other aspects of multidisciplinary strategies are in-depth summarized in the literature, while our mini-review pursues a different goal. From an ethical and medical standpoint, we witness a delayed implementation of novel therapies given the cost/time consuming process of organizing randomized trials that would bridge an already excellent local control in cT3-4 node-positive disease with long-term survival. This unfortunate separation of clinical research and medical care provides a strong motivation to repurpose known pharmaceuticals that suit for treatment intensification with a focus on distant control. In the framework of on-going phase II-III IG/IMRT-SIB trials, we came across an intriguing translational observation that the ratio of circulating (protumor) myeloid-derived suppressor cells to (antitumor) central memory CD8+ T cells is drastically increased, a possible mechanism of tumor immuno-escape and spread. This finding prompts that restoring the CD45RO memory T-cell pool could be a part of integrated adjuvant interventions. Therefore, the immunocorrective potentials of modified IL-2 and the anti-diabetic drug metformin are thoroughly discussed in the context of tumor immunobiology, mTOR pathways and revised Warburg effect.
- Published
- 2018
- Full Text
- View/download PDF
31. Immune checkpoint blockade combined with IL-6 and TGF-β inhibition improves the therapeutic outcome of mRNA-based immunotherapy
- Author
-
Dries Renmans, Kris Thielemans, Kevin Van der Jeught, Lukasz Bialkowski, Joeri L. Aerts, Sanne Bevers, Patrick Tjok Joe, and Carlo Heirman
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,business.industry ,medicine.drug_class ,medicine.medical_treatment ,Immunotherapy ,Monoclonal antibody ,Immune checkpoint ,Proinflammatory cytokine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,Cancer stem cell ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,business ,Cancer immunology - Abstract
Improved understanding of cancer immunology has provided insight into the phenomenon of frequent tumor recurrence after initially successful immunotherapy. A delicate balance exists between the capacity of the immune system to control tumor growth and various resistance mechanisms that arise to avoid or even counteract the host's immune system. These resistance mechanisms include but are not limited to (i) adaptive expression of inhibitory checkpoint molecules in response to the proinflammatory environment and (ii) amplification of cancer stem cells, a small fraction of tumor cells possessing the capacity for self-renewal and mediating treatment resistance and formation of metastases after long periods of clinical remission. Several individual therapeutic agents have so far been developed to revert T-cell exhaustion or disrupt the cross-talk between cancer stem cells and the tumor-promoting microenvironment. Here, we demonstrate that a three-arm combination therapy-consisting of an mRNA-based vaccine to induce antigen-specific T-cell responses, monoclonal antibodies blocking inhibitory checkpoint molecules (PD-1, TIM-3, LAG-3), and antibodies targeting IL-6 and TGF-β-improves the therapeutic outcome in subcutaneous TC-1 tumors and significantly prolongs survival of treated mice. Our findings point to a need for a rational development of multidimensional anticancer therapies, aiming at the induction of tumor-specific immunity and simultaneously targeting multiple resistance mechanisms.
- Published
- 2018
- Full Text
- View/download PDF
32. Intranodal administration of mRNA encoding nucleoprotein provides cross-strain immunity against influenza in mice
- Author
-
Joeri L. Aerts, Carlo Heirman, Patrick Tjok Joe, Xavier Saelens, Kris Thielemans, Lien Van Hoecke, Tine Ysenbaert, Ioanna Christopoulou, Bert Schepens, Faculty of Economic and Social Sciences and Solvay Business School, Faculty of Medicine and Pharmacy, Laboratory of Molecullar and Cellular Therapy, Basic (bio-) Medical Sciences, Hematology, Immunomodulation in Chronic Inflammatory Diseases, Medical Oncology, Physiology, and Pharmaceutical and Pharmacological Sciences
- Subjects
0301 basic medicine ,DNA VACCINATION ,T-Lymphocytes ,PROTECTIVE EFFICACY ,lcsh:Medicine ,Antibodies, Viral ,Bronchoalveolar Lavage ,Madin Darby Canine Kidney Cells ,Mice ,Intranodal ,0302 clinical medicine ,PLASMID DNA ,Medicine and Health Sciences ,A VIRUS-INFECTION ,PRECLINICAL EVALUATION ,Mice, Inbred BALB C ,Viral Vaccine ,General Medicine ,3. Good health ,Vaccination ,Influenza Vaccines ,030220 oncology & carcinogenesis ,Female ,Plasmids ,Influenza vaccine ,mRNA ,CD8(+) T-CELLS ,Biology ,DENDRITIC CELLS ,General Biochemistry, Genetics and Molecular Biology ,Virus ,DNA vaccination ,Interferon-gamma ,03 medical and health sciences ,Dogs ,Immune system ,Orthomyxoviridae Infections ,Antigen ,Immunity ,VACCINES ,Animals ,Humans ,RNA, Messenger ,Antigens ,Nucleoprotein ,Research ,Influenza A Virus, H3N2 Subtype ,lcsh:R ,RECOGNITION ,Biology and Life Sciences ,T cell ,Influenza ,Nucleoproteins ,Universal vaccine ,030104 developmental biology ,Immunology ,INJECTION - Abstract
Background Current human influenza vaccines lack the adaptability to match the mutational rate of the virus and therefore require annual revisions. Because of extensive manufacturing times and the possibility that antigenic alterations occur during viral vaccine strain production, an inherent risk exists for antigenic mismatch between the new influenza vaccine and circulating viruses. Targeting more conserved antigens such as nucleoprotein (NP) could provide a more sustainable vaccination strategy by inducing long term and heterosubtypic protection against influenza. We previously demonstrated that intranodal mRNA injection can induce potent antigen-specific T-cell responses. In this study, we investigated whether intranodal administration of mRNA encoding NP can induce T-cell responses capable of protecting against a heterologous influenza virus challenge. Methods BALB/c mice were immunized in the inguinal lymph nodes with different vaccination regimens of mRNA encoding NP. Immune responses were compared with NP DNA vaccination via IFN-γ ELISPOT and in vivo cytotoxicity. For survival experiments, mice were prime-boost vaccinated with 17 µg NP mRNA and infected with 1LD50 of H1N1 influenza virus 8 weeks after boost. Weight was monitored and viral titers, cytokines and immune cell populations in the bronchoalveolar lavage, and IFN-γ responses in the spleen were analyzed. Results Our results demonstrate that NP mRNA induces superior systemic T-cell responses against NP compared to classical DNA vaccination. These responses were sustained for several weeks even at low vaccine doses. Upon challenge infection, vaccination with NP mRNA resulted in reduced lung viral titers and improved recovery from infection. Finally, we show that vaccination with NP mRNA affects the immune response in infected lungs by lowering immune cell infiltration while increasing the fraction of T cells, monocytes and MHC II+ alveolar macrophages within immune infiltrates. This change was associated with altered levels of both pro- and anti-inflammatory cytokines. Conclusions These findings suggest that intranodal vaccination with NP mRNA induces cross-strain immunity against influenza, but also highlight a paradox of influenza immunity, whereby robust immune responses can provide protection, but can also transiently exacerbate symptoms during infection. Electronic supplementary material The online version of this article (10.1186/s12967-019-1991-3) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
33. Comparative analysis of antibodies to xCT (Slc7a11): Forewarned is forearmed
- Author
-
Ilse Julia Smolders, Thomas Demuyser, Ann Massie, Eduard Bentea, Ellen Merckx, Silvia Holmseth, Niels C. Danbolt, Joeri L. Aerts, Giulia Albertini, Lutgarde Arckens, Hideyo Sato, Joeri Van Liefferinge, and Virginie Follin-Arbelet
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,biology ,medicine.diagnostic_test ,General Neuroscience ,Glutamate receptor ,SLC7A11 ,Molecular biology ,Blot ,03 medical and health sciences ,Immunolabeling ,030104 developmental biology ,0302 clinical medicine ,Western blot ,Antigen ,Forebrain ,biology.protein ,medicine ,Immunohistochemistry ,030217 neurology & neurosurgery - Abstract
The cystine/glutamate antiporter or system Xc- exchanges cystine for glutamate, thereby supporting intracellular glutathione synthesis and nonvesicular glutamate release. The role of system Xc- in neurological disorders can be dual and remains a matter of debate. One important reason for the contradictory findings that have been reported to date is the use of nonspecific anti-xCT (the specific subunit of system Xc-) antibodies. Often studies rely on the predicted molecular weight of 55.5 kDa to identify xCT on Western blots. However, using brain extracts from xCT knockout (xCT(-/-)) mice as negative controls, we show that xCT migrates as a 35-kDa protein. Misinterpretation of immunoblots leads to incorrect assessment of antibody specificity and thereby to erroneous data interpretation. Here we have verified the specificity of most commonly used commercial and some in-house-developed anti-xCT antibodies by comparing their immunoreactivity in brain tissue of xCT(+/+) and xCT(-/-) mice by Western blotting and immunohistochemistry. The Western blot screening results demonstrate that antibody specificity not only differs between batches produced by immunizing different rabbits with the same antigen but also between bleedings of the same rabbit. Moreover, distinct immunohistochemical protocols have been tested for all the anti-xCT antibodies that were specific on Western blots in order to obtain a specific immunolabeling. Only one of our in-house-developed antibodies could reveal specific xCT labeling and exclusively on acetone-postfixed cryosections. Using this approach, we observed xCT protein expression throughout the mouse forebrain, including cortex, striatum, hippocampus, midbrain, thalamus, and amygdala, with greatest expression in regions facing the cerebrospinal fluid and meninges.
- Published
- 2015
- Full Text
- View/download PDF
34. DC immunotherapy in HIV-1 infection induces a major blood transcriptome shift
- Author
-
Carel A. van Baalen, Henk-Jan van den Ham, Arno C. Andeweg, Maarten Bijl, Wilfred F. J. van IJcken, Patrick Lacor, Kris Thielemans, Joeri L. Aerts, Albert D. M. E. Osterhaus, Anna L. de Goede, Rob A. Gruters, Sabine Allard, Brenda De Keersmaecker, Fatiha Zaaraoui-Boutahar, Sofie Wilgenhof, Marchina E. van der Ende, Pharmacy, Virology, Cell biology, Faculty of Medicine and Pharmacy, Laboratory of Molecullar and Cellular Therapy, Basic (bio-) Medical Sciences, and Clinical sciences
- Subjects
Adult ,Male ,Cellular immunity ,Microarray ,Anti-HIV Agents ,medicine.medical_treatment ,HIV Infections ,Biology ,Cancer Vaccines ,Transcriptome ,Immune system ,SDG 3 - Good Health and Well-being ,medicine ,Humans ,Melanoma ,AIDS Vaccines ,Inflammation ,Immunity, Cellular ,Principal Component Analysis ,General Veterinary ,General Immunology and Microbiology ,Vaccination ,Public Health, Environmental and Occupational Health ,Dendritic Cells ,Immunotherapy ,Middle Aged ,Viral Load ,Gene expression profiling ,Infectious Diseases ,lnfectious Diseases and Global Health Radboud Institute for Health Sciences [Radboudumc 4] ,Influenza Vaccines ,Immunology ,HIV-1 ,Leukocytes, Mononuclear ,Molecular Medicine ,Female ,Viral load - Abstract
Contains fulltext : 154919.pdf (Publisher’s version ) (Closed access) OBJECTIVE: This study aimed to evaluate the effect of dendritic cell (DC) vaccination against HIV-1 on host gene expression profiles. DESIGN: Longitudinal PBMC samples were collected from participants of the DC-TRN trial for immunotherapy against HIV. Microarray-assisted gene expression profiling was performed to evaluate the effects of vaccination and subsequent interruption of antiretroviral therapy on host genome expression. Data from the DC-TRN trial were compared with results from other vaccination trials. METHODS: We used Affymetrix GeneChips for microarray gene expression analysis. Data were analyzed by principal component analysis and differential gene expression was assessed using linear modeling. Gene ontology enrichment and gene set analysis were used to characterize differentially expressed genes. Transcriptome analysis included comparison with PBMCs obtained from DC-vaccinated melanoma patients and of healthy individuals who received seasonal influenza vaccination. RESULTS: DC-TRN immunotherapy in HIV-infected individuals resulted in a major shift in the transcriptome. Longitudinal analysis demonstrated that changes in the transcriptome sustained also during interruption of antiretroviral therapy. After DC-vaccination, the transcriptome was enriched for cellular immunity associated genes that were also induced in healthy adults who received live attenuated influenza virus vaccination. These beneficial responses were accompanied by detrimental signals of general immune activation. CONCLUSIONS: The DC-TRN induced changes in the transcriptome were profound, lasting, and consisted of both protective signals and signatures of inflammation and immune exhaustion, with a net result of decreased viral load, without clinical benefit. Thus transcriptome analysis provides useful information, dissecting both positive and negative effects, for the evaluation of safety and efficacy of immunotherapeutic strategies.
- Published
- 2015
- Full Text
- View/download PDF
35. Immune checkpoint blockade combined with IL-6 and TGF-β inhibition improves the therapeutic outcome of mRNA-based immunotherapy
- Author
-
Lukasz, Bialkowski, Kevin, Van der Jeught, Sanne, Bevers, Patrick, Tjok Joe, Dries, Renmans, Carlo, Heirman, Joeri L, Aerts, and Kris, Thielemans
- Subjects
Interleukin-6 ,SOXB1 Transcription Factors ,Melanoma, Experimental ,Gene Expression ,Disease Models, Animal ,Mice ,Antineoplastic Agents, Immunological ,Recurrence ,Transforming Growth Factor beta ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Immunotherapy ,RNA, Messenger ,Signal Transduction - Abstract
Improved understanding of cancer immunology has provided insight into the phenomenon of frequent tumor recurrence after initially successful immunotherapy. A delicate balance exists between the capacity of the immune system to control tumor growth and various resistance mechanisms that arise to avoid or even counteract the host's immune system. These resistance mechanisms include but are not limited to (i) adaptive expression of inhibitory checkpoint molecules in response to the proinflammatory environment and (ii) amplification of cancer stem cells, a small fraction of tumor cells possessing the capacity for self-renewal and mediating treatment resistance and formation of metastases after long periods of clinical remission. Several individual therapeutic agents have so far been developed to revert T-cell exhaustion or disrupt the cross-talk between cancer stem cells and the tumor-promoting microenvironment. Here, we demonstrate that a three-arm combination therapy-consisting of an mRNA-based vaccine to induce antigen-specific T-cell responses, monoclonal antibodies blocking inhibitory checkpoint molecules (PD-1, TIM-3, LAG-3), and antibodies targeting IL-6 and TGF-β-improves the therapeutic outcome in subcutaneous TC-1 tumors and significantly prolongs survival of treated mice. Our findings point to a need for a rational development of multidimensional anticancer therapies, aiming at the induction of tumor-specific immunity and simultaneously targeting multiple resistance mechanisms.
- Published
- 2017
36. Oncolytic virus-induced cell death and immunity: a match made in heaven?
- Author
-
Iain A. McNeish, Jolien De Munck, Joeri L. Aerts, Alex Binks, Faculty of Sciences and Bioengineering Sciences, Pharmaceutical and Pharmacological Sciences, Faculty of Medicine and Pharmacy, Laboratory of Molecullar and Cellular Therapy, and Faculty of Economic and Social Sciences and Solvay Business School
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Review ,03 medical and health sciences ,Immune system ,Neoplasms ,Journal Article ,Immunology and Allergy ,Medicine ,Animals ,Humans ,Virotherapy ,Oncolytic Virotherapy ,business.industry ,Cancer ,Cell Biology ,Immunotherapy ,medicine.disease ,Immune checkpoint ,Oncolytic virus ,Radiation therapy ,Oncolytic Viruses ,030104 developmental biology ,cell death ,Cancer research ,Immunogenic cell death ,business - Abstract
Our understanding of the mechanisms responsible for cancer development has increased enormously over the last decades. However, for many cancers, this has not been translated into a significant improvement in overall survival, and overall mortality remains high. Treatment for many malignancies remains based on surgery, chemotherapy, and radiotherapy. Significant progress has been made toward the development of more specific, more potent, and less invasive treatment modalities, but such targeted therapies remain the exception for most cancers. Thus, cancer therapies based on a different mechanism of action should be explored. The immune system plays an important role in keeping tumor growth at bay. However, in many cases, these responses are not strong enough to keep tumor growth under control. Thus, immunotherapy aims to boost the immune system to suppress tumor growth efficiently. This has been demonstrated by the recent successes of immune checkpoint therapy in several cancers. Oncolytic viruses (OVs) are another exciting class of immunotherapy agent. As well as replicating selectively within and killing tumor cells, OVs are able to elicit potent anti-tumor immune responses. Therapeutic vaccination with OVs, also referred to as cancer virotherapy, can thus be tailored to elicit vigorous cellular immune responses and even target individual malignancies in a personalized manner. In this review, we will describe the intricate link among oncolytic virotherapy, tumor immunology, and immunogenic cell death (ICD) and discuss ways to harness optimally their potential for future cancer therapy.
- Published
- 2017
37. Immunomodulatory drugs improve the immune environment for dendritic cell-based immunotherapy in multiple myeloma patients after autologous stem cell transplantation
- Author
-
Karel Fostier, Brenda De Keersmaecker, Sofie Wilgenhof, Carlo Heirman, Jurgen Corthals, Rik Schots, Joeri L. Aerts, Kris Thielemans, Laboratory of Molecullar and Cellular Therapy, Faculty of Medicine and Pharmacy, Basic (bio-) Medical Sciences, Immunology and Microbiology, Clinical sciences, and Hematology
- Subjects
Adult ,Male ,Cancer Research ,Transplantation Conditioning ,medicine.medical_treatment ,Immunology ,pomalidomide ,Angiogenesis Inhibitors ,Immunotherapy, Adoptive ,Transplantation, Autologous ,Dexamethasone ,Immunomodulation ,Immune system ,Autologous stem-cell transplantation ,immunomodulatory drugs ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Immunologic Factors ,Immunology and Allergy ,Cytotoxic T cell ,Lenalidomide ,Melphalan ,Multiple myeloma ,Cell Proliferation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Dendritic Cells ,Dendritic cell ,Immunotherapy ,Middle Aged ,medicine.disease ,Thalidomide ,Transplantation ,medicine.anatomical_structure ,Oncology ,Doxorubicin ,Vincristine ,Female ,immunotherapy ,Bone marrow ,Multiple Myeloma ,business - Abstract
Multiple myeloma (MM) is characterized by a malignant proliferation of plasma cells in the bone marrow with associated organ damage. Although the prognosis of MM has improved recently, the disease remains incurable for the large majority of patients. The eradication of residual disease in the bone marrow is a main target on the road toward cure. Immune cells play a role in the control of cancer and can be tools to attack residual MM cells. However, the myeloma-associated immune deficiency is a major hurdle to immunotherapy. We evaluated ex vivo the effects of low doses of the immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide on several immune cell types from MM patients after autologous stem cell transplantation and with low tumor burden. We observed that these drugs increased CD4(+) and CD8(+) T-cell proliferation and cytokine production, enhanced the lytic capacity of cytotoxic T lymphocytes and reduced the suppressive effects of regulatory T cells on CD8(+) T-cell responses. In addition, we found that functional dendritic cells (DCs) can be generated from mononuclear cells from MM patients. The presence of IMiDs improved the quality of antigen-specific T cells induced or expanded by these DCs as evidenced by a higher degree of T-cell polyfunctionality. Our results provide a rationale for the design of early phase clinical studies to assess the efficacy of DC-based immunotherapy in combination with posttransplant maintenance treatment with IMiDs in MM.
- Published
- 2014
- Full Text
- View/download PDF
38. Phosphorylated STAT3 physically interacts with NPM and transcriptionally enhances its expression in cancer
- Author
-
Karine Breckpot, Z Ren, Joeri L. Aerts, J. P. De Greve, Carlo Heirman, Joeri Pen, Laboratory of Molecullar and Cellular Therapy, Internal Medicine Specializations, Medical Imaging and Physical Sciences, Basic (bio-) Medical Sciences, Clinical sciences, Laboratory of Molecular and Medical Oncology, Physiology, and Pathology/molecular and cellular medicine
- Subjects
STAT3 Transcription Factor ,Cancer Research ,Transcription, Genetic ,NPM ,Biology ,medicine.disease_cause ,Jurkat cells ,STAT3 ,Cell Line, Tumor ,Neoplasms ,Genetics ,medicine ,Humans ,Anaplastic lymphoma kinase ,Phosphorylation ,Molecular Biology ,Cancer ,Regulation of gene expression ,integumentary system ,Interferon-alpha ,Nuclear Proteins ,phosphorylated ,Gene Expression Regulation, Neoplastic ,Protein Transport ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Cancer cell ,Cancer research ,STAT protein ,Carcinogenesis ,Nucleophosmin ,Tyrosine kinase ,Signal Transduction - Abstract
The signal transducer and activator of transcription 3 (STAT3) can be activated by the tyrosine kinase domain of the chimeric protein nucleophosmin/anaplastic lymphoma kinase (NPM/ALK), and has a pivotal role in mediating NPM/ALK-related malignant cell transformation. Although the role of STAT3 and wild-type NPM in oncogenesis has been extensively investigated, the relationship between both molecules in cancer remains poorly understood. In the present study, we first demonstrate that STAT3 phosphorylation at tyrosine 705 is accompanied by a concomitant increase in the expression level of NPM. Nuclear co-translocation of phosphorylated STAT3 with NPM can be triggered by interferon-alpha (IFN-a) stimulation of Jurkat cells and phosphorylated STAT3 co-localizes with NPM in cancer cells showing constitutive STAT3 activation. We further demonstrate that STAT3 phosphorylation can transcriptionally mediate NPM upregulation in IFN-a-stimulated Jurkat cells and is responsible for maintaining its expression in cancer cells showing constitutive STAT3 activation. Inhibition of STAT3 phosphorylation or knockdown of NPM expression abrogates their simultaneous transnuclear movements. Finally, we found evidence for a physical interaction between NPM and STAT3 in conditions of STAT3 activation. In conclusion, NPM is a downstream effector of the STAT3 signaling, and can facilitate the nuclear entry of phosphorylated STAT3. These observations might open novel opportunities for targeting the STAT3 pathway in cancer.
- Published
- 2014
- Full Text
- View/download PDF
39. Enhanced suppressive capacity of tumor‐infiltrating myeloid‐derived suppressor cells compared with their peripheral counterparts
- Author
-
Perpetua Emeagi, Kris Thielemans, Sarah Maenhout, Sandra Van Lint, and Joeri L. Aerts
- Subjects
Cancer Research ,Myeloid ,Regulatory T cell ,Spleen ,Biology ,Nitric Oxide ,T-Lymphocytes, Regulatory ,Mice ,Downregulation and upregulation ,Cell Line, Tumor ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Myeloid Cells ,Tumor microenvironment ,Arginase ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oncology ,Immunology ,B7-1 Antigen ,Myeloid-derived Suppressor Cell ,Female ,CD80 ,CD8 - Abstract
Although the main site of action for myeloid-derived suppressor cells (MDSCs) is most likely the tumor microenvironment, so far the study of these cells has been largely restricted to spleen-derived MDSCs. In this study, we compared the suppressive capacity of splenic and tumor-derived MDSCs in different subcutaneous mouse tumor models. We investigated which suppressive mechanisms were involved. Finally, we investigated whether MDSCs and regulatory T cells (Treg ) cooperate in the suppression of T-cell responses. In all models, splenic granulocytic MDSCs (grMDSC) strongly suppress CD4(+) T-cell proliferation while the suppressive effect on CD8(+) T cells is less pronounced. Splenic monocytic MDSCs (moMDSC) have a lower suppressive capacity, compared to grMDSC, on both CD4(+) and CD8(+) T-cell proliferation. Both grMDSC and moMDSC isolated from the tumor have a much stronger suppressive activity compared to MDSCs isolated from the spleen of tumor-bearing mice, associated with a higher NO2 (-) production by the tumor-derived moMDSC and arginase activity for both subsets. The expression of CD80 is also elevated on tumor-derived grMDSC compared with their peripheral counterparts. Direct contact with tumor cells is required for the upregulation of CD80 and CD80(+) MDSCs are more suppressive than CD80(-) MDSCs. Coculture of Treg and MDSCs leads to a stronger suppression of CD8(+) T-cell proliferation compared to the suppression observed by Treg or MDSCs alone. Thus, we showed that tumor-infiltrating MDSCs possess a stronger suppressive capacity than their peripheral counterparts and that various suppressive mechanisms account for this difference.
- Published
- 2013
- Full Text
- View/download PDF
40. Phosphorylated STAT5 regulates p53 expression via BRCA1/BARD1-NPM1 and MDM2
- Author
-
Jack P Chen, Hugo Vandenplas, Vuk Stambolic, Zhuo Ren, Jiance A Wang, Mark D. Minden, Jacques De Greve, Eldad Zacksenhaus, Cleo Goyvaerts, Carlo Heirman, Olena Gorbenko, Philippe Giron, Joeri L. Aerts, Karine Breckpot, Laboratory for Medical and Molecular Oncology, Faculty of Medicine and Pharmacy, Laboratory of Molecullar and Cellular Therapy, Pharmaceutical and Pharmacological Sciences, Clinical sciences, and Basic (bio-) Medical Sciences
- Subjects
0301 basic medicine ,Cancer Research ,Cell Survival ,Ubiquitin-Protein Ligases ,Immunology ,Down-Regulation ,Models, Biological ,Cell Line ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Downregulation and upregulation ,hemic and lymphatic diseases ,STAT5 Transcription Factor ,Humans ,Phosphorylation ,Phosphotyrosine ,STAT5 ,Nucleophosmin ,biology ,Chemistry ,BRCA1 Protein ,Tumor Suppressor Proteins ,Nuclear Proteins ,food and beverages ,Proto-Oncogene Proteins c-mdm2 ,Cell Biology ,Ubiquitin ligase ,030104 developmental biology ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Proteolysis ,biology.protein ,STAT protein ,Cancer research ,Mdm2 ,Original Article ,Signal transduction ,Tumor Suppressor Protein p53 ,Protein Binding - Abstract
Signal transducer and activator of transcription 5 (STAT5) and nucleophosmin (NPM1) are critical regulators of multiple biological and pathological processes. Although a reciprocal regulatory relationship was established between STAT5A and a NPM–ALK fusion protein in T-cell lymphoma, no direct connection between STAT5 and wild-type NPM1 has been documented. Here we demonstrate a mutually regulatory relationship between STAT5 and NPM1. Induction of STAT5 phosphorylation at Y694 (P-STAT5) diminished NPM1 expression, whereas inhibition of STAT5 phosphorylation enhanced NPM1 expression. Conversely, NPM1 not only negatively regulated STAT5 phosphorylation but also preserved unphosphorylated STAT5 level. Mechanistically, we show that NPM1 downregulation by P-STAT5 is mediated by impairing the BRCA1-BARD1 ubiquitin ligase, which controls the stability of NPM1. In turn, decreased NPM1 levels led to suppression of p53 expression, resulting in enhanced cell survival. This study reveals a new STAT5 signaling pathway regulating p53 expression via NPM1 and uncovers new therapeutic targets for anticancer treatment in tumors driven by STAT5 signaling.
- Published
- 2016
- Full Text
- View/download PDF
41. Disease progression in recurrent glioblastoma patients treated with the VEGFR inhibitor axitinib is associated with increased regulatory T cell numbers and T cell exhaustion
- Author
-
Daphné Benteyn, Johnny Duerinck, Bart Neyns, Kris Thielemans, Stephanie Du Four, Joeri L. Aerts, Brenda De Keersmaecker, Sarah Maenhout, Clinical sciences, Faculty of Medicine and Pharmacy, Laboratory of Molecullar and Cellular Therapy, Basic (bio-) Medical Sciences, Neuroprotection & Neuromodulation, Physiology, Immunomodulation in Chronic Inflammatory Diseases, Laboratory of Molecular and Medical Oncology, and Pharmaceutical and Pharmacological Sciences
- Subjects
Male ,0301 basic medicine ,increased regulatory ,Cancer Research ,Axitinib ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Angiogenesis Inhibitors ,VEGFR inhibitor axitinib ,T-Lymphocytes, Regulatory ,0302 clinical medicine ,Immunophenotyping ,T-Lymphocyte Subsets ,Immunology and Allergy ,Hepatitis A Virus Cellular Receptor 2 ,T cell exhaustion ,Imidazoles ,Middle Aged ,Lymphocyte Activation Gene 3 Protein ,Phenotype ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Disease Progression ,Cytokines ,Female ,medicine.drug ,Adult ,Indazoles ,Regulatory T cell ,T cell ,Immunology ,Immunomodulation ,03 medical and health sciences ,Immune system ,Antigens, CD ,diseaese porgression ,Cell Line, Tumor ,medicine ,Humans ,Lymphocyte Count ,Protein Kinase Inhibitors ,Aged ,Tumor microenvironment ,business.industry ,Immunotherapy ,Receptors, Vascular Endothelial Growth Factor ,030104 developmental biology ,Cancer research ,Neoplasm Recurrence, Local ,Glioblastoma ,business ,Immunologic Memory ,Biomarkers ,CD8 - Abstract
BACKGROUND: Recurrent glioblastoma is associated with a poor overall survival. Antiangiogenic therapy results in a high tumor response rate but has limited impact on survival. Immunotherapy has emerged as an efficient treatment modality for some cancers, and preclinical evidence indicates that anti-VEGF(R) therapy can counterbalance the immunosuppressive tumor microenvironment. METHODS: We collected peripheral blood mononuclear cells (PBMC) of patients with recurrent glioblastoma treated in a randomized phase II clinical trial comparing the effect of axitinib with axitinib plus lomustine and analyzed the immunophenotype of PBMC, the production of cytokines and expression of inhibitory molecules by circulating T cells. RESULTS: PBMC of 18 patients were collected at baseline and at 6 weeks after initiation of study treatment. Axitinib increased the number of naïve CD8(+) T cells and central memory CD4(+) and CD8(+) T cells and reduced the TIM3 expression on CD4(+) and CD8(+) T cells. Patients diagnosed with progressive disease on axitinib had a significantly increased number of regulatory T cells and an increased level of PD-1 expression on CD4(+) and CD8(+) T cells. In addition, reduced numbers of cytokine-producing T cells were found in progressive patients as compared to patients responding to treatment. CONCLUSION: Our results suggest that axitinib treatment in patients with recurrent glioblastoma has a favorable impact on immune function. At the time of acquired resistance to axitinib, we documented further enhancement of a preexisting immunosuppression. Further investigations on the role of axitinib as potential combination partner with immunotherapy are necessary.
- Published
- 2016
42. Combined VEGFR and CTLA-4 blockade increases the antigen-presenting function of intratumoral DCs and reduces the suppressive capacity of intratumoral MDSCs
- Author
-
Stephanie, Du Four, Sarah K, Maenhout, Simone P, Niclou, Kris, Thielemans, Bart, Neyns, and Joeri L, Aerts
- Subjects
Original Article - Abstract
Melanoma brain metastases (MBM) occur in 10% to 50% of melanoma patients. They are often associated with a high morbidity and despite the improvements in the treatment of advanced melanoma, including immunotherapy, patients with MBM still have a poor prognosis. Antiangiogenic treatment was shown to reduce the immunosuppressive tumor microenvironment. Therefore we investigated the effect of the combination of VEGFR- and CTLA-4 blockade on the immune cells within the tumor microenvironment. In this study we investigated the effect of the combination of axitinib, a TKI against VEGFR-1, -2 and -3, with therapeutic inhibition of CTLA-4 in subcutaneous and intracranial mouse melanoma models. The combination of axitinib with αCTLA-4 reduced tumor growth and increased survival in both intracranial and subcutaneous models. Investigation of the splenic immune cells showed an increased number of CD4+ and CD8+ T cells after combination treatment. Moreover, combination treatment increased the number of intratumoral dendritic cells (DCs) and monocytic myeloid-derived suppressor cells (moMDSCs). When these immune cell populations were sorted from the subcutaneous and intracranial tumors of mice treated with axitinib+αCTLA-4, we observed an increased antigen-presenting function of DCs and a reduced suppressive capacity of moMDSCs on a per cell basis. Our results suggest that the combination of antiangiogenesis and checkpoint inhibition can lead to an enhanced antitumor effect leading to increased survival. We found that this effect is in part due to an enhanced antitumor immune response generated by an increased antigen-presenting function of intratumoral DCs in combination with a reduced suppressive capacity of intratumoral moMDSCs.
- Published
- 2016
43. Intralymphatic mRNA vaccine induces CD8 T-cell responses that inhibit the growth of mucosally located tumours
- Author
-
Dries Renmans, Kris Thielemans, Joeri L. Aerts, Geert Stangé, Kevin Van der Jeught, Lukasz Bialkowski, Lidia Daszkiewicz, Carlo Heirman, Alexia van Weijnen, Karine Breckpot, Basic (bio-) Medical Sciences, Laboratory of Molecullar and Cellular Therapy, Faculty of Medicine and Pharmacy, Medicine and Pharmacy academic/administration, Pharmaceutical and Pharmacological Sciences, Physiology, Immunomodulation in Chronic Inflammatory Diseases, Diabetes Pathology & Therapy, and Microbiology and Infection Control
- Subjects
0301 basic medicine ,Uterine Cervical Neoplasms ,CD8-Positive T-Lymphocytes ,Biology ,Cancer Vaccines ,Article ,CD49a ,Mice ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Cytotoxic T cell ,RNA, Messenger ,Cervical cancer ,Cisplatin ,Vaccines ,Tumor microenvironment ,Mucous Membrane ,Multidisciplinary ,Mucous membrane ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Female ,Immunologic Memory ,CD8 ,medicine.drug - Abstract
The lack of appropriate mouse models is likely one of the reasons of a limited translational success rate of therapeutic vaccines against cervical cancer, as rapidly growing ectopic tumours are commonly used for preclinical studies. In this work, we demonstrate that the tumour microenvironment of TC-1 tumours differs significantly depending on the anatomical location of tumour lesions (i.e. subcutaneously, in the lungs and in the genital tract). Our data demonstrate that E7-TriMix mRNA vaccine-induced CD8+ T lymphocytes migrate into the tumour nest and control tumour growth, although they do not express mucosa-associated markers such as CD103 or CD49a. We additionally show that despite the presence of the antigen-specific T cells in the tumour lesions, the therapeutic outcomes in the genital tract model remain limited. Here, we report that such a hostile tumour microenvironment can be reversed by cisplatin treatment, leading to a complete regression of clinically relevant tumours when combined with mRNA immunization. We thereby demonstrate the necessity of utilizing clinically relevant models for preclinical evaluation of anticancer therapies and the importance of a simultaneous combination of anticancer immune response induction with targeting of tumour environment.
- Published
- 2016
- Full Text
- View/download PDF
44. Comparative analysis of antibodies to xCT (Slc7a11): Forewarned is forearmed
- Author
-
Joeri, Van Liefferinge, Eduard, Bentea, Thomas, Demuyser, Giulia, Albertini, Virginie, Follin-Arbelet, Silvia, Holmseth, Ellen, Merckx, Hideyo, Sato, Joeri L, Aerts, Ilse, Smolders, Lutgarde, Arckens, Niels C, Danbolt, and Ann, Massie
- Subjects
Male ,Mice, Knockout ,Amino Acid Transport System y+ ,Molecular Sequence Data ,Brain ,Sequence Homology ,Rats ,Mice ,Sequence Analysis, Protein ,Animals ,Humans ,Amino Acid Sequence ,Rabbits ,Autoantibodies - Abstract
The cystine/glutamate antiporter or system Xc- exchanges cystine for glutamate, thereby supporting intracellular glutathione synthesis and nonvesicular glutamate release. The role of system Xc- in neurological disorders can be dual and remains a matter of debate. One important reason for the contradictory findings that have been reported to date is the use of nonspecific anti-xCT (the specific subunit of system Xc-) antibodies. Often studies rely on the predicted molecular weight of 55.5 kDa to identify xCT on Western blots. However, using brain extracts from xCT knockout (xCT(-/-)) mice as negative controls, we show that xCT migrates as a 35-kDa protein. Misinterpretation of immunoblots leads to incorrect assessment of antibody specificity and thereby to erroneous data interpretation. Here we have verified the specificity of most commonly used commercial and some in-house-developed anti-xCT antibodies by comparing their immunoreactivity in brain tissue of xCT(+/+) and xCT(-/-) mice by Western blotting and immunohistochemistry. The Western blot screening results demonstrate that antibody specificity not only differs between batches produced by immunizing different rabbits with the same antigen but also between bleedings of the same rabbit. Moreover, distinct immunohistochemical protocols have been tested for all the anti-xCT antibodies that were specific on Western blots in order to obtain a specific immunolabeling. Only one of our in-house-developed antibodies could reveal specific xCT labeling and exclusively on acetone-postfixed cryosections. Using this approach, we observed xCT protein expression throughout the mouse forebrain, including cortex, striatum, hippocampus, midbrain, thalamus, and amygdala, with greatest expression in regions facing the cerebrospinal fluid and meninges.
- Published
- 2015
45. The Interferon Inducer Ampligen [Poly(I)-Poly(C 12 U)] Markedly Protects Mice against Coxsackie B3 Virus-Induced Myocarditis
- Author
-
Armando M. De Palma, Elizaveta Padalko, Dieter Nuyens, Johan Neyts, Erik De Clercq, Joeri L. Aerts, Peter Carmeliet, Erik Verbeken, Pharmaceutical and Pharmacological Sciences, Laboratory of Molecullar and Cellular Therapy, and Pathologic Biochemistry and Physiology
- Subjects
Myocardium/pathology ,Male ,Poly U ,Interferon Inducers ,Myocarditis ,Viral Myocarditis ,Heart/virology ,Interferon Inducers/therapeutic use ,Coxsackievirus Infections ,Alpha interferon ,Interferon alpha-2 ,Biology ,Antiviral Agents ,Poly I-C/therapeutic use ,Virus ,Cercopithecus aethiops ,Electrocardiography ,Mice ,Interferon ,Chlorocebus aethiops ,medicine ,Animals ,Pharmacology (medical) ,Vero Cells ,Antiviral Agents/therapeutic use ,Pharmacology ,Mice, Inbred C3H ,Interferon inducer ,Poly U/therapeutic use ,Reverse Transcriptase Polymerase Chain Reaction ,Myocardium ,Interferon-alpha/therapeutic use ,Interferon-alpha ,Heart ,medicine.disease ,Virology ,Recombinant Proteins ,Myocarditis/pathology ,Enterovirus B, Human ,Coxsackievirus Infections/prevention & control ,Titer ,Poly I-C ,Infectious Diseases ,RNA, Viral/analysis ,Vero cell ,RNA, Viral ,medicine.drug - Abstract
Viral replication, as well as an immunopathological component, is assumed to be involved in coxsackie B virus-induced myocarditis. We evaluated the efficacy of the interferon inducer Ampligen on coxsackie B3 virus-induced myocarditis in C3H/HeNHsd mice. The efficacy of Ampligen was compared with that of the interferon inducer poly(inosinic acid)-poly(cytidylic acid) [poly(IC)], alpha interferon 2b (INTRON A), and pegylated alpha interferon 2b (PEG-INTRON-α-2b). Ampligen at 20 mg/kg of body weight/day was able to reduce the severity of virus-induced myocarditis, as assessed by morphometric analysis, by 98% ( P = 3.0 × 10 −8 ). When poly(IC) was administered at 15 mg/kg/day, it reduced the severity of virus-induced myocarditis by 93% ( P = 5.6 × 10 −5 ). Alpha interferon 2b (1 × 10 5 U/day) and pegylated alpha interferon 2b (5 × 10 5 U/day) were less effective and reduced the severity of virus-induced myocarditis by 66% ( P = 0.0009) and 78% ( P = 0.0002), respectively. The observed efficacies of Ampligen and poly(IC) were corroborated by the observation that the drugs also markedly reduced the virus titers in the heart, as detected by (i) quantitative real-time reverse transcription-PCR and (ii) titration for infectious virus content. Whereas the electrocardiograms for untreated mice with myocarditis were severely disturbed, the electrocardiographic parameters were normalized in Ampligen- and poly(IC)-treated mice. Even when start of treatment with Ampligen was delayed until day 2 postinfection, a time at which lesions had already appeared in untreated control animals, a marked protective effect on the development of viral myocarditis (as assessed at day 6 postinfection) was still noted.
- Published
- 2004
- Full Text
- View/download PDF
46. [Untitled]
- Author
-
Philippe Vandekerckhove and Joeri L. Aerts
- Subjects
Mrna expression ,Cytokine expression ,Bioengineering ,Quantitative Reverse Transcriptase PCR ,General Medicine ,Biology ,Applied Microbiology and Biotechnology ,Molecular biology ,Real-time polymerase chain reaction ,Healthy volunteers ,Receptor ,Tumor necrosis factor receptor ,Biotechnology ,Transforming growth factor - Abstract
We have applied and validated a recently described method (real-time quantitative PCR) for cytokine mRNA quantification. This method is easy to optimise and offers sensitive, reliable and quantitative results with significantly reduced labour, allowing the simultaneous analysis of cytokine expression for large numbers of samples. With this method the expression levels of several cytokines (TNF-α and TGF-β1) and their receptors (FAS, TNFR1 and 2) have been compared between eosinophilic and neutrophilic granulocytes isolated from peripheral blood. No differences were found using the healthy volunteer samples.
- Published
- 2002
- Full Text
- View/download PDF
47. Location, location, location: functional and phenotypic heterogeneity between tumor-infiltrating and non-infiltrating myeloid-derived suppressor cells
- Author
-
Joeri L. Aerts, Kris Thielemans, Sarah Maenhout, Basic (bio-) Medical Sciences, Laboratory of Molecullar and Cellular Therapy, Physiology, and Immunomodulation in Chronic Inflammatory Diseases
- Subjects
Tumor microenvironment ,Immunology ,Cancer ,Spleen ,Review ,Biology ,medicine.disease ,Phenotype ,law.invention ,medicine.anatomical_structure ,Lymphatic system ,Immune system ,Oncology ,law ,medicine ,Myeloid-derived Suppressor Cell ,Immunology and Allergy ,Suppressor - Abstract
An increasing number of studies is focusing on the role of myeloid-derived suppressor cells (MDSCs) in the suppression of antitumor immune responses. Although the main site of action for MDSCs is most likely the tumor microenvironment, the study of these cells has been largely restricted to MDSCs derived from peripheral lymphoid organs. Only in a minority of studies MDSCs isolated from the tumor microenvironment have been characterized. This review will give an overview of the data available on the phenotypical and functional differences between tumor-derived MDSCs and MDSCs isolated from the spleen of tumor-bearing mice or from the peripheral blood of cancer patients.
- Published
- 2014
48. AZD1480 delays tumor growth in a melanoma model while enhancing the suppressive activity of myeloid-derived suppressor cells
- Author
-
Stephanie Du Four, Sarah Maenhout, Joeri L. Aerts, Bart Neyns, Kris Thielemans, Jurgen Corthals, Internal Medicine Specializations, Laboratory of Molecullar and Cellular Therapy, Clinical sciences, Laboratory of Molecular and Medical Oncology, Physiology, and Basic (bio-) Medical Sciences
- Subjects
tumor ,Myeloid ,Melanoma, Experimental ,Biology ,JAK1/2 inhibitors ,Mice ,Random Allocation ,Immune system ,Cell Line, Tumor ,medicine ,melanoma ,Animals ,Humans ,Myeloid Cells ,tumor immunology ,STAT3 ,Cell Proliferation ,immunosuppression ,Cell growth ,Melanoma ,suppressor cells ,JAK-STAT signaling pathway ,Janus Kinase 2 ,myeloid-derived suppressor cells ,medicine.disease ,Xenograft Model Antitumor Assays ,AZD1480 ,Mice, Inbred C57BL ,Disease Models, Animal ,Pyrimidines ,medicine.anatomical_structure ,Oncology ,Cell culture ,Immunology ,signal transducer and activator of transcription 3 ,Myeloid-derived Suppressor Cell ,Cancer research ,biology.protein ,Pyrazoles ,Female ,myeloid ,Signal Transduction ,Research Paper - Abstract
AZD1480 is a potent, competitive small-molecule inhibitor of JAK1/2 kinase which inhibits STAT3 phosphorylation and tumor growth. Here we investigated the effects of AZD1480 on the function of different immune cell populations in a melanoma model. When MO4 tumor-bearing mice were treated with AZD1480 we observed a strong inhibition of tumor growth as well as a prolonged survival. Moreover, a significant decrease in the percentage of myeloid-derived suppressor cells (MDSCs) was observed after treatment with AZD1480. However, AZD1480 enhanced the suppressive capacity of murine MDSCs while at the same time impairing the proliferative as well as the IFN-γ secretion capacity of murine T cells. The addition of AZD1480 to co-cultures of human MDSCs and T cells does not affect the suppressive activity of MDSCs but it does reduce the IFN-γ secretion and the proliferative capacity of T cells. We showed that although AZD1480 has the ability to delay the tumor growth of MO4 tumor-bearing mice, this drug has detrimental effects on several aspects of the immune system. These data indicate that systemic targeting of the JAK/STAT pathway by JAK1/2 inhibition can have divergent effects on tumor growth and anti-tumor immune responses.
- Published
- 2014
- Full Text
- View/download PDF
49. Manipulating Immune Regulatory Pathways to Enhance T Cell Stimulation
- Author
-
Therese Liechtenstein, David Escors, Joeri J. Pen, Joeri L. Aerts, and Karine Breckpot
- Subjects
medicine.medical_treatment ,T cell ,Cancer ,chemical and pharmacologic phenomena ,Stimulation ,Biology ,medicine.disease ,medicine.anatomical_structure ,Immune system ,Cancer immunotherapy ,Immunity ,Cancer research ,medicine ,Cytotoxic T cell ,CD70 - Abstract
Cancer immunotherapy aspires to treat malignant disease by activating cancer specific immune responses. It is generally accepted that the latter can only be achieved by an approach in which tumor specific T cells are educated to recognize and kill tumor cells, whilst they are furthermore empowered to overcome immunosuppressive mechanisms present both at peripheral sites and in the tumor environment. Dendritic cells (DCs) have been extensively explored as a cellular vaccine for the stimulation of tumor specific T cells. Several strategies have been devised to manipulate these cells to become strongly activated tumor associated antigen (TAA) presenting cells. Our growing knowledge on the biology of DCs and the costimulatory as well as inhibitory molecules expressed by them, provides us with opportunities to generate DCs that are capable of hyper-activating cytotoxic T lymphocytes (CTLs) whilst they impact on regulatory T cells (Treg), which are now well established to be an important contributor to failure of cancer vaccines. In this chapter, we will focus on the cross talk between DCs and T cells mediated by the CD70/CD27 and PD-L1/PD-1 axis as these have been identified as critical pathways in the regulation of immunity versus tolerance.
- Published
- 2014
- Full Text
- View/download PDF
50. β2-adrenergic agonists modulate TNF-α induced astrocytic inflammatory gene expression and brain inflammatory cell populations
- Author
-
Joeri L. Aerts, Jacques De Keyser, Guy Laureys, Anneleen Spooren, Frauke Demol, and Sarah Gerlo
- Subjects
Male ,NF-KAPPA-B ,NF-κB ,DISEASE ,ACTIVATION ,chemistry.chemical_compound ,Neuroinflammation ,Medicine and Health Sciences ,Receptor ,Cells, Cultured ,IN-VIVO ,NEUTROPHILS ,TUMOR-NECROSIS-FACTOR ,β2-adrenergic receptors ,General Neuroscience ,Intracellular Signaling Peptides and Proteins ,NF-kappa B ,Brain ,MULTIPLE-SCLEROSIS ,Intercellular Adhesion Molecule-1 ,Cell biology ,DNA-Binding Proteins ,Cysteine Endopeptidases ,Crosstalk (biology) ,Neurology ,Cytokines ,Encephalitis ,Tumor necrosis factor alpha ,medicine.symptom ,Signal transduction ,SPINAL-CORD ,Ubiquitin-Protein Ligases ,Immunology ,Vascular Cell Adhesion Molecule-1 ,Inflammation ,Astrocytoma ,Biology ,beta(2)-adrenergic receptors ,Cellular and Molecular Neuroscience ,medicine ,Animals ,Humans ,Clenbuterol ,REGULATORY T-CELLS ,Rats, Wistar ,Adrenergic beta-2 Receptor Agonists ,Tumor Necrosis Factor alpha-Induced Protein 3 ,Tumor Necrosis Factor-alpha ,Research ,CENTRAL-NERVOUS-SYSTEM ,NFKB1 ,Rats ,Disease Models, Animal ,Animals, Newborn ,chemistry ,Astrocytes ,Neuroscience - Abstract
Background: The NF-kappa B signaling pathway orchestrates many of the intricate aspects of neuroinflammation. Astrocytic beta(2)-adrenergic receptors have emerged as potential regulators in central nervous system inflammation and are potential targets for pharmacological modulation. The aim of this study was to elucidate the crosstalk between astrocytic beta(2)-adrenergic receptors and the TNF-alpha induced inflammatory gene program.Methods: Proinflammatory conditions were generated by the administration of TNF-alpha. Genes that are susceptible to astrocytic crosstalk between beta(2)-adrenergic receptors (stimulated by clenbuterol) and TNF-alpha were identified by qPCR-macroarray-based gene expression analysis in a human 1321 N1 astrocytoma cell line. Transcriptional patterns of the identified genes in vitro were validated by RT-PCR on the 1321 N1 cell line as well as on primary rat astrocytes. In vivo expression patterns were examined by intracerebroventricular administration of clenbuterol and/or TNF-alpha in rats. To examine the impact on the inflammatory cell content of the brain we performed extensive FACS analysis of rat brain immune cells after intracerebroventricular clenbuterol and/or TNF-alpha administration.Results: Parallel transcriptional patterns in vivo and in vitro confirmed the relevance of astrocytic beta(2)-adrenergic receptors as modulators of brain inflammatory responses. Importantly, we observed pronounced effects of beta(2)-adrenergic receptor agonists and TNF-alpha on IL-6, CXCL2, CXCL3, VCAM1, and ICAM1 expression, suggesting a role in inflammatory brain cell homeostasis. Extensive FACS-analysis of inflammatory cell content in the brain demonstrated that clenbuterol/TNF-alpha co-administration skewed the T cell population towards a double negative phenotype and induced a shift in the myeloid brain cell population towards a neutrophilic predominance.Conclusions: Our results show that astrocytic beta(2)-adrenergic receptors are potent regulators of astrocytic TNF-alpha-activated genes in vitro and in vivo, and ultimately modulate the molecular network involved in the homeostasis of inflammatory cells in the central nervous system. Astrocytic beta(2)-adrenergic receptors and their downstream signaling pathway may serve as potential targets to modulate neuroinflammatory responses.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.