96 results on '"Corthals J"'
Search Results
2. A phase IB study on intravenous synthetic mRNA electroporated dendritic cell immunotherapy in pretreated advanced melanoma patients
- Author
-
Wilgenhof, S., Van Nuffel, A.M.T., Benteyn, D., Corthals, J., Aerts, C., Heirman, C., Van Riet, I., Bonehill, A., Thielemans, K., and Neyns, B.
- Published
- 2013
- Full Text
- View/download PDF
3. Interference with PD-L1/PD-1 co-stimulation during antigen presentation enhances the multifunctionality of antigen-specific T cells
- Author
-
Pen, J J, Keersmaecker, B D, Heirman, C, Corthals, J, Liechtenstein, T, Escors, D, Thielemans, K, and Breckpot, K
- Published
- 2014
- Full Text
- View/download PDF
4. Induction of antigen-specific CD8+ cytotoxic T cells by dendritic cells co-electroporated with a dsRNA analogue and tumor antigen mRNA
- Author
-
Michiels, A, Breckpot, K, Corthals, J, Tuyaerts, S, Bonehill, A, Heirman, C, Thielemans, K, and Aerts, J L
- Published
- 2006
- Full Text
- View/download PDF
5. Electroporation of immature and mature dendritic cells: implications for dendritic cell-based vaccines
- Author
-
Michiels, A, Tuyaerts, S, Bonehill, A, Corthals, J, Breckpot, K, Heirman, C, Van Meirvenne, S, Dullaers, M, Allard, S, Brasseur, F, van der Bruggen, P, and Thielemans, K
- Published
- 2005
- Full Text
- View/download PDF
6. Loading of dendritic cells for immunotherapy
- Author
-
Van Nuffel, A. M. T., Benteyn, D., Wilgenhof, S., Corthals, J., Heirman, C., Neyns, B., Bonehill, A., and Thielemans, K.
- Published
- 2013
- Full Text
- View/download PDF
7. Moran, P.: De origine Scoticae linguae (O’Mulconry’s glossary)…
- Author
-
Corthals, J., primary
- Published
- 2020
- Full Text
- View/download PDF
8. A MAGE-3 peptide presented by HLA-B44 is also recognized by cytolytic T lymphocytes on HLA-B18
- Author
-
Bilsborough, J., Panichelli, C., Duffour, M-T., Warnier, G., Lurquin, C., Schultz, E. S., Thielemans, K., Corthals, J., Boon, T., and Bruggen, P. van der
- Published
- 2002
9. Corrigendum to 'A phase I/IIa immunotherapy trial of HIV-1-infected patients with Tat, Rev and Nef expressing dendritic cells followed by treatment interruption (vol 142, pg 252, 2012)
- Author
-
Allard, SD, De Keersmaecker, B, Goede, Anna, Verschuren, Esther, Koetsveld, Jeanette, Reedijk, Marleen, Wylock, C, De Bel, AV, Vandeloo, J, Pistoor, FHM (Frank), Heirman, C, Beyer, Walter, Eilers, Paul, Corthals, J, Padmos, I, Thielemans, K, Osterhaus, Ab, Lacor, P, Ende, Marchina, Aerts, JL, Baalen, Carel, Gruters, Rob, Pharmacy, Virology, Epidemiology, and Internal Medicine
- Subjects
SDG 3 - Good Health and Well-being - Published
- 2014
10. 3328 A randomized controlled phase II clinical trial on autologous monocyte-derived mRNA electroporated dendritic cells for stage III/IV melanoma patients who are disease-free following the local treatment of macrometastases
- Author
-
Jansen, Y., primary, Corthals, J., additional, Wilgenhof, S., additional, Schreuer, M., additional, Heirman, C., additional, Thielemans, K., additional, and Neyns, B., additional
- Published
- 2015
- Full Text
- View/download PDF
11. Dendritic cell-based immune therapy against HIV-1
- Author
-
Gruters, R. A., de Keersmaecker, B., de Goede, A. L., Allard, S. D., Koetsveld, J., Corthals, J., Schutten, M., Heirman, C., van der Ende, M. E., Lacor, P., Osterhaus, A. D., Thielemans, K., van Baalen, C. A., Aerts, Joeri, Physiology, Laboratory of Molecullar and Cellular Therapy, Clinical sciences, Faculty of Medicine and Pharmacy, Pharmaceutical and Pharmacological Sciences, and Microbiology and Infection Control
- Abstract
HIV-1 infected patients on HAART were treated with mRNA electroporated autologous dendritic cells (DC) in a phase I/II clinical trial and the effect of active immune therapy on immune responses and viral sequence evolution was evaluated. HIV patients (n=17) were vaccinated with DC expressing Tat, Rev and Nef. After four vaccinations HAART treatment was interrupted. PBMC taken during and after vaccination were screened with Elispot, using either peptides or electroporated DC for restimulation. For sequence analysis, HIV RNA was amplified by RT-PCR; sequence variation in vaccine and control genes was analyzed pre- and post-vaccination. In 12 out of 16 patients screened with both peptide and DC Elispot, increased post-vaccination responses to vaccine-antigens were found. In 12/17 patients a complete set of both pre-HAART and post-vaccination sequences was obtained. With one exception, variation in pre-HAART and post vaccination HIV sequences was limited. Viral sequences spanning specific HLA restricted epitopes showed evidence of viral evolution, but this was not a common phenomenon. The immune therapy was well-tolerated and did not have adverse effects. Increased cellular immunity against the antigens could be demonstrated. The enhanced immunity resulted in higher sequence variability in vaccine genes than control genes, although the effects were limited. There was no significant correlation between the breadth and/or extent of the immune response and the rate of virus evolution.
- Published
- 2009
12. P18-03. Dendritic cell-based immune therapy against HIV-1
- Author
-
Gruters, R.A. (Rob), Keersmaecker, B. (Brenda) de, Goede, A.L. (Anna) de, Allard, S.D., Koetsveld, J. (Jeanette), Corthals, J. (Jurgen), Schutten, M. (Martin), Heirman, C. (Carlo), Ende, M.E. (Marchina) van der, Lacor, P. (Patrick), Osterhaus, A.D.M.E. (Albert), Thielemans, K. (Kris), Baalen, C.A. (Carel) van, Aerts, J.L. (Joeri), Gruters, R.A. (Rob), Keersmaecker, B. (Brenda) de, Goede, A.L. (Anna) de, Allard, S.D., Koetsveld, J. (Jeanette), Corthals, J. (Jurgen), Schutten, M. (Martin), Heirman, C. (Carlo), Ende, M.E. (Marchina) van der, Lacor, P. (Patrick), Osterhaus, A.D.M.E. (Albert), Thielemans, K. (Kris), Baalen, C.A. (Carel) van, and Aerts, J.L. (Joeri)
- Published
- 2009
- Full Text
- View/download PDF
13. Electroporation of immature and mature dendritic cells: implications for dendritic cell-based vaccines
- Author
-
UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, UCL - MD/MINT - Département de médecine interne, UCL - (SLuc) Unité d'oncologie médicale, Michiels, A., Tuyaerts, Sandra, Bonehill, A, Corthals, J, Breckpot, K, Heirman, C., Van Meirvenne, S, Dullaers, M, Allard, S, Brasseur, Francis, van der Bruggen, Pierre, Thielemans, K., UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, UCL - MD/MINT - Département de médecine interne, UCL - (SLuc) Unité d'oncologie médicale, Michiels, A., Tuyaerts, Sandra, Bonehill, A, Corthals, J, Breckpot, K, Heirman, C., Van Meirvenne, S, Dullaers, M, Allard, S, Brasseur, Francis, van der Bruggen, Pierre, and Thielemans, K.
- Abstract
Until now, studies utilizing mRNA electroporation as a tool for the delivery of tumor antigens to human monocyte-derived dendritic cells ( DC) have focused on DC electroporated in an immature state. Immature DC are considered to be specialized in antigen capture and processing, whereas mature DC present antigen and have an increased T-cell stimulatory capacity. Therefore, the consensus has been to electroporate DC before maturation. We show that the transfection efficiency of DC electroporated either before or after maturation was similarly high. Both immature and mature electroporated DC, matured in the presence of an inflammatory cytokine cocktail, expressed mature DC surface markers and preserved their capacity to secrete cytokines and chemokines upon CD40 ligation. In addition, both immature and mature DC can be efficiently cryopreserved before or after electroporation without deleterious effects on viability, phenotype or T-cell stimulatory capacity including in vitro antigen-specific T-cell activation. However, DC electroporated after maturation are more efficient in in vitro migration assays and at least as effective in antigen presentation as DC electroporated before maturation. These results are important for vaccination strategies where an optimal antigen presentation by DC after migration to the lymphoid organs is crucial.
- Published
- 2005
14. A phase I clinical trial on the combined intravenous (IV) and intradermal (ID) administration of autologous TriMix-DC cellular therapy in patients with pretreated melanoma (TriMixIDIV).
- Author
-
Neyns, B., primary, Wilgenhof, S., additional, Van Nuffel, A. M. T., additional, Benteyn, D., additional, Heirman, C., additional, Van Riet, I., additional, Bonehill, A., additional, Corthals, J., additional, and Thielemans, K., additional
- Published
- 2011
- Full Text
- View/download PDF
15. A MAGE-3 peptide presented by HLA-DR1 to CD4(+) T cells that were isolated from a melanoma patient vaccinated with a MAGE-3 protein
- Author
-
UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, Zhang, Y., Chaux, P, Stroobant, Vincent, Eggermont, AMM, Corthals, J, Maillere, B, Thielemans, K., Marchand, Marie, Boon, Thierry, van der Bruggen, Pierre, UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, Zhang, Y., Chaux, P, Stroobant, Vincent, Eggermont, AMM, Corthals, J, Maillere, B, Thielemans, K., Marchand, Marie, Boon, Thierry, and van der Bruggen, Pierre
- Abstract
"Cancer-germline" genes such as those of the MAGE family are expressed in many tumors and in male germline cells, but are silent in normal tissues. They encode shared tumor-specific Ags, which have been used in therapeutic vaccination trials of cancer patients. MAGE-3 is expressed in 74% of metastatic melanoma and in 50% of carcinomas of esophagus, head and neck, bladder, and lung. We report here the identification of a new MAGE-3 peptide, which is recognized by three different CD4(+) T cell clones isolated from a melanoma patient vaccinated with a MAGE-3 protein. These clones, which express different TCRs, recognize on HLA-DR1 peptide ACYEFLWGPRALVETS, which corresponds to the MAGE-3(267-282), and the MAGE-12(267)-(282) protein sequences. One of the T cell clones, which expresses LFA-1 at a high level, lysed tumor cells expressing DR1 and MAGE-3. Another of these DR1-restricted CD4(+) clones recognized not only the MAGE-3/12 peptide but also homologous peptides encoded by genes MAGE-1, 2, 4, 6, 10, and 11.
- Published
- 2003
16. FC18 Clinical outcome following therapeutic vaccination with autologous mRNA electroporated dendritic cell vaccines in patients with advanced melanoma
- Author
-
Pierret, L., primary, Neyns, B., additional, Wilgenhof, S., additional, Lienard, D., additional, De Coninck, A., additional, Corthals, J., additional, Bonehill, A., additional, Roseeuw, D., additional, del Marmol, V., additional, and Thielemans, K., additional
- Published
- 2010
- Full Text
- View/download PDF
17. Clinical outcome following therapeutic vaccination with autologous mRNA electroporated dendritic cell (DC) vaccines in patients with advanced melanoma.
- Author
-
Wilgenhof, S., primary, Van Nuffel, A. M., additional, Benteyn, D., additional, Corthals, J., additional, Heirman, C., additional, Pierret, L., additional, De Coninck, A., additional, Bonehill, A., additional, Thielemans, K., additional, and Neyns, B., additional
- Published
- 2010
- Full Text
- View/download PDF
18. A MAGE-3 peptide presented by HLA-B44 is also recognized by cytolytic T lymphocytes on HLA-B18
- Author
-
UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, Bilsborough, Janine, Panichelli, C, Duffour, MT, Warnier, G., Lurquin, C., Schultz, ES, Thielemans, K., Corthals, J, Boon, Thierry, van der Bruggen, Pierre, UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, Bilsborough, Janine, Panichelli, C, Duffour, MT, Warnier, G., Lurquin, C., Schultz, ES, Thielemans, K., Corthals, J, Boon, Thierry, and van der Bruggen, Pierre
- Abstract
Antigens encoded by MAGE genes are of particular interest for cancer immunotherapy because of their tumoral specificity and because they are shared by many tumors. Antigenic peptide MEVDPIGHLY, which is encoded by MAGE-3 and is known to be presented by human leukocyte antigen (HLA)-B44, is currently being used in therapeutic vaccination trials. We report here that a cytolytic T lymphocyte (CTL) clone, which is restricted by HLA-B*1801, recognizes the same peptide and, importantly, lyzes HLA-B18 tumor cells expressing MAGE-3. These results imply that the use of peptide MEVDPIGHLY can now be extended to HLA-B18 patients. We also provide evidence that, under limiting amounts of protein MAGE-3, HLA B*1801 and B*4403 compete for binding to the peptide.
- Published
- 2002
19. P18-03. Dendritic cell-based immune therapy against HIV-1
- Author
-
Gruters, RA, primary, de Keersmaecker, B, additional, de Goede, AL, additional, Allard, SD, additional, Koetsveld, J, additional, Corthals, J, additional, Schutten, M, additional, Heirman, C, additional, Ende, ME van der, additional, Lacor, P, additional, Osterhaus, AD, additional, Thielemans, K, additional, van Baalen, CA, additional, and Aerts, JL, additional
- Published
- 2009
- Full Text
- View/download PDF
20. Correlation between prior therapeutic dendritic cell vaccination and the outcome of patients with metastatic melanoma treated with ipilimumab
- Author
-
Pierret, L., primary, Wilgenhof, S., additional, Corthals, J., additional, Roelandt, T., additional, Thielemans, K., additional, and Neyns, B., additional
- Published
- 2009
- Full Text
- View/download PDF
21. A MAGE-1 peptide recognized on HLA-DR15 by CD4(+) T cells.
- Author
-
UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, Chaux, P, Lethé, B, Van Snick, Jacques, Corthals, J, Schultz, E S, Cambiaso, Cesar, Boon, Thierry, van der Bruggen, Pierre, UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, Chaux, P, Lethé, B, Van Snick, Jacques, Corthals, J, Schultz, E S, Cambiaso, Cesar, Boon, Thierry, and van der Bruggen, Pierre
- Abstract
Antigens encoded by MAGE genes and recognized by T cells are of interest for cancer immunotherapy because of their strict tumoral specificity and because they are shared by many tumors. Several MAGE-1 peptide that are recognized by CD8(+) cytolytic T lymphocytes have been used in therapeutic vaccination trials. To obtain anti-tumor immune response, vaccines combining peptides recognized by CD8(+) and peptides recognized by CD4(+) T cells might be optimal. We focused therefore on the identification of MAGE peptides recognized by CD4(+) T cells. We report here the identification of MAGE-1 epitope EYVIKVSARVRF, which is presented to CD4(+) T lymphocytes by HLA-DR15. This HLA allele is present in 29 % of Asians and 17 % of Caucasians.
- Published
- 2001
22. Interferon alfa-2b treatment following therapeutic vaccination with mRNA electroporated dendritic cells results in skin depigmentation and tumor regression in patients with advanced melanoma
- Author
-
Neyns, B., primary, Corthals, J., additional, and Thielemans, K., additional
- Published
- 2007
- Full Text
- View/download PDF
23. A cohort pilot study on therapeutic vaccination of advanced melanoma patients with dendritic cells loaded with multiple peptides or electroporated with mRNA
- Author
-
Neyns, B., primary, Corthals, J., additional, De Grève, J., additional, De Greef, C., additional, Coulie, P. G., additional, and Thielemans, K., additional
- Published
- 2006
- Full Text
- View/download PDF
24. IRSLINGER, B. S.: Abstrakta mit Dentalsuffixen im Altirischen
- Author
-
Corthals, J., primary
- Published
- 2005
- Full Text
- View/download PDF
25. Die Trennung von Finn und Gráinne
- Author
-
CORTHALS, J., primary
- Published
- 1997
- Full Text
- View/download PDF
26. Loading of dendritic cells for immunotherapy.
- Author
-
Nuffel, A. M. T., Benteyn, D., Wilgenhof, S., Corthals, J., Heirman, C., Neyns, B., Bonehill, A., and Thielemans, K.
- Subjects
DENDRITIC cells ,IMMUNOTHERAPY ,IMMUNE response ,EPITOPES ,ANTIGENS ,T cells ,HLA histocompatibility antigens ,CLINICAL trials - Abstract
Dendritic cell therapy has been optimized a lot aiming to induce a strong and broad immune response in terms of the recognized epitopes by both CD8
+ and CD4+ T cells and the use of the patients' complete unique set of HLA molecules. We here give an overview of our approach for antigen loading and maturation of dendritic cells and describe the consequences to evaluate the immune response after treatment as well as the Brussels experience in clinical trials. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
27. Induction of antigen-specific CD8+ cytotoxic T cells by dendritic cells co-electroporated with a dsRNA analogue and tumor antigen mRNA.
- Author
-
Michiels, A, Breckpot, K, Corthals, J, Tuyaerts, S, Bonehill, A, Heirman, C, Thielemans, K, and Aerts, J L
- Subjects
DENDRITIC cells ,DOUBLE-stranded RNA ,ELECTROPORATION ,T cells ,IMMUNOTHERAPY ,TUMOR antigens - Abstract
The maturation state of dendritic cells (DCs) is an important determinant for the initiation and regulation of adaptive immune responses. In this study, we wanted to assess whether functional activation of human monocyte-derived DCs can be achieved by electroporation of an activation signal in the form of double-stranded (ds) RNA and whether simultaneous electroporation of the dsRNA with tumor antigen encoding mRNA can lead to the induction of a cytotoxic T-lymphocyte (CTL) response. Electroporation of immature DCs with poly(I:C
12 U), a dsRNA analogue, resulted in phenotypic as well as functional changes, indicative of DC maturation. Co-electroporation of DCs with both poly(I:C12 U) and Melan-A/MART-1 encoding mRNA induced strong anti-Melan-A/MART-1 CD8+ T-cell responses in vitro. Higher numbers of Melan-A/MART-1-specific CTLs were consistently obtained with poly(I:C12 U)-activated DCs compared to DCs matured in the presence of an inflammatory cytokine cocktail. These results indicate that DC co-electroporation with both dsRNA and tumor antigen encoding mRNA induces fully activated and antigen-loaded DCs that promote antigen-specific CTL responses and may provide the basis for future immunotherapeutic strategies.Gene Therapy (2006) 13, 1027–1036. doi:10.1038/sj.gt.3302750; published online 2 March 2006 [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
28. A randomized controlled phase II clinical trial on mRNA electroporated autologous monocyte-derived dendritic cells (TriMixDC-MEL) as adjuvant treatment for stage III/IV melanoma patients who are disease-free following the resection of macrometastases.
- Author
-
Jansen Y, Kruse V, Corthals J, Schats K, van Dam PJ, Seremet T, Heirman C, Brochez L, Kockx M, Thielemans K, and Neyns B
- Subjects
- Adult, Aged, Aged, 80 and over, CD27 Ligand genetics, CD27 Ligand immunology, CD40 Ligand genetics, CD40 Ligand immunology, Combined Modality Therapy methods, Dendritic Cells metabolism, Disease-Free Survival, Electroporation, Female, Follow-Up Studies, Humans, Immunotherapy methods, Male, Melanoma immunology, Melanoma mortality, Melanoma secondary, Middle Aged, Neoplasm Recurrence, Local prevention & control, Neoplasm Staging, RNA, Messenger genetics, Skin Neoplasms immunology, Skin Neoplasms mortality, Skin Neoplasms pathology, Surgical Procedures, Operative, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 immunology, Transplantation, Autologous methods, Young Adult, Dendritic Cells transplantation, Melanoma therapy, Neoplasm Recurrence, Local epidemiology, RNA, Messenger immunology, Skin Neoplasms therapy
- Abstract
Background: Autologous monocyte-derived mRNA co-electroporated dendritic cells with mRNA encoding CD40 ligand (CD40L), CD70 and a constitutively activated TLR4 (caTLR4) (referred to as TriMixDC-MEL) have anti-tumor activity in advanced melanoma patients. We investigated the safety and activity of adjuvant TriMixDC-MEL in stage III/IV melanoma patients., Materials and Methods: Forty-one patients were randomly assigned to treatment with TriMixDC-MEL (n = 21) and standard follow-up (n = 20). "Cross-over" was allowed at the time of non-salvageable recurrence. The primary endpoint was the percentage of patients alive and disease-free at 1-year. For a subset of patients, (formalin-fixed paraffin-embedded), tumor tissue samples were available for mRNA expression profiling and PD-L1 immunohistochemical staining., Results: Baseline characteristics were well balanced. One-year after randomization, 71% of patients in the study arm were alive and free of disease compared to 35% in the control arm. After a median follow-up of 53 months (range 3-67), 23 patients experienced a non-salvageable melanoma recurrence (TriMixDC-Mel arm n = 9 and control arm n = 14).The median time to non-salvageable recurrence was superior in the TriMixDC-MEL arm (median 8 months (range 1-6) vs. not reached; log-rank p 0.044). TriMixDC-MEL-related adverse events (AE) consisted of transient local skin reactions, flu-like symptoms and post-infusion chills. No grade ≥ 3 AE's occurred. The mRNA expression profiling revealed four genes (STAT2, TPSAB1, CD9 and CSF2) as potential predictive biomarkers., Conclusion: TriMixDC-MEL id/iv as adjuvant therapy is tolerable and may improve the 1-year disease-free survival rate. Combination of optimized autologous monocyte-derived DC-formulations warrants further investigation in combination with currently approved adjuvant therapy options.
- Published
- 2020
- Full Text
- View/download PDF
29. TriMix and tumor antigen mRNA electroporated dendritic cell vaccination plus ipilimumab: link between T-cell activation and clinical responses in advanced melanoma.
- Author
-
De Keersmaecker B, Claerhout S, Carrasco J, Bar I, Corthals J, Wilgenhof S, Neyns B, and Thielemans K
- Subjects
- Adult, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, Cancer Vaccines genetics, Cancer Vaccines immunology, Dendritic Cells immunology, Electroporation, Female, Follow-Up Studies, Humans, Immunization, Secondary, Lymphocyte Activation, Male, Melanoma diagnosis, Melanoma immunology, Melanoma mortality, Middle Aged, Neoplasm Staging, Progression-Free Survival, RNA, Messenger genetics, RNA, Messenger immunology, Remission Induction methods, Skin Neoplasms diagnosis, Skin Neoplasms immunology, Skin Neoplasms mortality, Transplantation, Autologous, Vaccination methods, Cancer Vaccines administration & dosage, Dendritic Cells transplantation, Ipilimumab administration & dosage, Melanoma therapy, Skin Neoplasms therapy
- Abstract
Background: We previously reported that dendritic cell-based mRNA vaccination plus ipilimumab (TriMixDC-MEL IPI) results in an encouraging rate of tumor responses in patients with pretreated advanced melanoma. Here, we report the TriMixDC-MEL IPI-induced T-cell responses detected in the peripheral blood., Methods: Monocyte-derived dendritic cells electroporated with mRNA encoding CD70, CD40 ligand, and constitutively active TLR4 (TriMix) as well as the tumor-associated antigens tyrosinase, gp100, MAGE-A3, or MAGE-C2 were administered together with IPI for four cycles. For 18/39 patients, an additional vaccine was administered before the first IPI administration. We evaluated tumor-associated antigen specific T-cell responses in previously collected peripheral blood mononuclear cells, available from 15 patients., Results: Vaccine-induced enzyme-linked immunospot assay responses detected after in vitro T-cell stimulation were shown in 12/15 patients. Immune responses detected in patients with a complete or partial response were significantly stronger and broader, and exhibited a higher degree of multifunctionality compared with responses in patients with stable or progressive disease. CD8+ T-cell responses from patients with an ongoing clinical response, either elicited by TriMixDC-MEL IPI or on subsequent pembrolizumab treatment, exhibited the highest degree of multifunctionality., Conclusions: TriMixDC-MEL IPI treatment results in robust CD8+ T-cell responses in a meaningful portion of stage III or IV melanoma patients, and obviously in patients with a clinical response. The levels of polyfunctional and multiantigen T-cell responses measured in patients with a complete response, particularly in patients evidently cured after 5+ years of follow-up, may provide a benchmark for the level of immune stimulation needed to achieve a durable clinical remission., Trial Registration Number: NCT01302496., Competing Interests: Competing interests: The use of dendritic cells electroporated with tumor antigen mRNA and TriMix is topic of a patent (W2009/034172) on which KT is filed as inventor. This patent has been licensed to eTheRNA immunotherapies. BDK and SC are respectively current and former employees of eTheRNA immunotherapies. BN has received financial compensation from Bristol-Myers Squibb for public speaking and participation in advisory board meetings., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
30. Single Domain Antibody-Mediated Blockade of Programmed Death-Ligand 1 on Dendritic Cells Enhances CD8 T-cell Activation and Cytokine Production.
- Author
-
Broos K, Lecocq Q, Keersmaecker B, Raes G, Corthals J, Lion E, Thielemans K, Devoogdt N, Keyaerts M, and Breckpot K
- Abstract
Dendritic cell [DC] vaccines can induce durable clinical responses, at least in a fraction of previously treated, late stage cancer patients. Several preclinical studies suggest that shielding programmed death-ligand 1 [PD-L1] on the DC surface may be an attractive strategy to extend such clinical benefits to a larger patient population. In this study, we evaluated the use of single domain antibody [sdAb] K2, a high affinity, antagonistic, PD-L1 specific sdAb, for its ability to enhance DC mediated T-cell activation and benchmarked it against the use of the monoclonal antibodies [mAbs], MIH1, 29E.2A3 and avelumab. Similar to mAbs, sdAb K2 enhanced antigen-specific T-cell receptor signaling in PD-1 positive (PD-1
pos ) reporter cells activated by DCs. We further showed that the activation and function of antigen-specific CD8 positive (CD8pos ) T cells, activated by DCs, was enhanced by inclusion of sdAb K2, but not mAbs. The failure of mAbs to enhance T-cell activation might be explained by their low efficacy to bind PD-L1 on DCs when compared to binding of PD-L1 on non-immune cells, whereas sdAb K2 shows high binding to PD-L1 on immune as well as non-immune cells. These data provide a rationale for the inclusion of sdAb K2 in DC-based immunotherapy strategies., Competing Interests: M.K. has received travel and accommodation expenses from Bayer NV and research funding from Camel-IDS; N.D. and G.R. are co-founders of Camel-IDs. N.D. has received funding from F. Hoffmann-La Roche, Telix Pharmaceuticals, Agenus, 121BIO, Complix and Boehringer Ingelheim.- Published
- 2019
- Full Text
- View/download PDF
31. Evaluating a Single Domain Antibody Targeting Human PD-L1 as a Nuclear Imaging and Therapeutic Agent.
- Author
-
Broos K, Lecocq Q, Xavier C, Bridoux J, Nguyen TT, Corthals J, Schoonooghe S, Lion E, Raes G, Keyaerts M, Devoogdt N, and Breckpot K
- Abstract
The PD-1:PD-L1 immune checkpoint axis is central in the escape of cancer cells from anticancer immune responses. Monoclonal antibodies (mAbs) specific for PD-L1 have been approved for treatment of various cancer types. Although PD-L1 blockade has proven its merit, there are still several aspects that require further attention to fully capitalize on its potential. One of these is the development of antigen-binding moieties that enable PD-L1 diagnosis and therapy. We generated human PD-L1 binding single domain antibodies (sdAbs) and selected sdAb K2, a sdAb with a high affinity for PD-L1, as a lead compound. SPECT/CT imaging in mice following intravenous injection of Technetium-99m (
99m Tc)-labeled sdAb K2 revealed high signal-to-noise ratios, strong ability to specifically detect PD-L1 in melanoma and breast tumors, and relatively low kidney retention, which is a unique property for radiolabeled sdAbs. We further showed using surface plasmon resonance that sdAb K2 binds to the same epitope on PD-L1 as the mAb avelumab, and antagonizes PD-1:PD-L1 interactions. Different human cell-based assays corroborated the PD-1:PD-L1 blocking activity, showing enhanced T-cell receptor signaling and tumor cell killing when PD-1POS T cells interacted with PD-L1POS tumor cells. Taken together, we present sdAb K2, which specifically binds to human PD-L1, as a new diagnostic and therapeutic agent in cancer management., Competing Interests: Supplementary Materials: The following are available online at www.mdpi.com/xxx/s1, Figure S1: Characteristics of PD-L1POS and PD-L1NEG 624-MEL and MCF7 tumors.- Published
- 2019
- Full Text
- View/download PDF
32. Impact of lenalidomide maintenance on the immune environment of multiple myeloma patients with low tumor burden after autologous stem cell transplantation.
- Author
-
Fostier K, Caers J, Meuleman N, Broos K, Corthals J, Thielemans K, Schots R, and De Keersmaecker B
- Abstract
Lenalidomide is a potent anti-myeloma drug with immunomodulatory properties. It is increasingly used in a low-dose maintenance setting to prolong remission duration after standard treatment. Data on the in vivo effects of lenalidomide are scarce and sometimes different from the well-described in vitro effects. We therefore evaluated the numerical, phenotypical and functional impact of lenalidomide maintenance on several immune cell types in a cohort of seventeen homogeneously treated myeloma patients achieving a low residual myeloma burden after a bortezomib based-induction followed by autologous stem cell transplantation. Lenalidomide maintenance: 1) increased the fraction of naïve CD8
+ T cells and several memory T-cell subsets, 2) reduced the numbers of terminal effector CD8+ T cells, 3) resulted in a higher expression of co-stimulatory molecules on resting T cells and of the inhibitory checkpoint molecules LAG-3 on CD4+ T cells and TIM-3 on CD4+ and CD8+ T cells, 4) reduced the number of TIGIT+ CD8+ T cells, 5) increased the number of regulatory T cells with a phenotype associated with strong suppressive capacity. Purified CD8+ T cells showed increased and more polyfunctional recall viral responses. However, PBMC responses were not enhanced during lenalidomide maintenance and CD4+ T-cell responses specific for the myeloma-associated antigen MAGE-C1 even tended to become lower. We conclude that lenalidomide maintenance after autologous stem cell transplantation has complex pleotropic effects on the immune environment. Immune interventions such as anti-myeloma vaccination should include measures to tackle an expanded inhibitory Treg compartment., Competing Interests: CONFLICTS OF INTEREST None.- Published
- 2018
- Full Text
- View/download PDF
33. Phase II Study of Autologous Monocyte-Derived mRNA Electroporated Dendritic Cells (TriMixDC-MEL) Plus Ipilimumab in Patients With Pretreated Advanced Melanoma.
- Author
-
Wilgenhof S, Corthals J, Heirman C, van Baren N, Lucas S, Kvistborg P, Thielemans K, and Neyns B
- Subjects
- Adult, Aged, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal adverse effects, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Cells, Cultured, Chemotherapy, Adjuvant, Dendritic Cells immunology, Dendritic Cells metabolism, Disease Progression, Disease-Free Survival, Drug Administration Schedule, Female, Genetic Therapy adverse effects, Genetic Therapy mortality, Humans, Ipilimumab, Kaplan-Meier Estimate, Male, Melanoma genetics, Melanoma immunology, Melanoma mortality, Middle Aged, Proportional Hazards Models, RNA, Messenger metabolism, Skin Neoplasms genetics, Skin Neoplasms immunology, Skin Neoplasms mortality, Time Factors, Transplantation, Autologous, Treatment Outcome, Young Adult, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Dendritic Cells transplantation, Electroporation, Genetic Therapy methods, Melanoma therapy, RNA, Messenger genetics, Skin Neoplasms therapy
- Abstract
Purpose: Autologous monocyte-derived dendritic cells (DCs) electroporated with synthetic mRNA (TriMixDC-MEL) are immunogenic and have antitumor activity as a monotherapy in patients with pretreated advanced melanoma. Ipilimumab, an immunoglobulin G1 monoclonal antibody directed against the cytotoxic T-lymphocyte-associated protein 4 receptor that counteracts physiologic suppression of T-cell function, improves the overall survival of patients with advanced melanoma. This phase II study investigated the combination of TriMixDC-MEL and ipilimumab in patients with pretreated advanced melanoma., Patients and Methods: Thirty-nine patients were treated with TriMixDC-MEL (4 × 10(6) cells administered intradermally and 20 × 10(6) cells administered intravenously) plus ipilimumab (10 mg/kg every 3 weeks for a total of four administrations, followed by maintenance therapy every 12 weeks in patients who remained progression free). Six-month disease control rate according to the immune-related response criteria served as the primary end point., Results: The 6-month disease control rate was 51% (95% CI, 36% to 67%), and the overall tumor response rate was 38% (including eight complete and seven partial responses). Seven complete responses and one partial tumor response are ongoing after a median follow-up time of 36 months (range, 22 to 43 months). The most common treatment-related adverse events (all grades) consisted of local DC injection site skin reactions (100%), transient post-DC infusion chills (38%) and flu-like symptoms (84%), dermatitis (64%), hepatitis (13%), hypophysitis (15%), and diarrhea/colitis (15%). Grade 3 or 4 immune-related adverse events occurred in 36% of patients. There was no grade 5 adverse event., Conclusion: The combination of TriMixDC-MEL and ipilimumab is tolerable and results in an encouraging rate of highly durable tumor responses in patients with pretreated advanced melanoma., (© 2016 by American Society of Clinical Oncology.)
- Published
- 2016
- Full Text
- View/download PDF
34. mRNA Electroporation of Dendritic Cells with WT1, Survivin, and TriMix (a Mixture of caTLR4, CD40L, and CD70).
- Author
-
Coosemans A, Tuyaerts S, Morias K, Corthals J, Heirman C, Thielemans K, Van Gool SW, Vergote I, and Amant F
- Subjects
- Antigens, Neoplasm genetics, CD27 Ligand genetics, CD27 Ligand immunology, CD40 Ligand genetics, CD40 Ligand immunology, Cells, Cultured, Dendritic Cells cytology, Humans, Inhibitor of Apoptosis Proteins genetics, Inhibitor of Apoptosis Proteins immunology, RNA, Messenger genetics, Survivin, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 immunology, WT1 Proteins genetics, WT1 Proteins immunology, Antigens, Neoplasm immunology, Dendritic Cells immunology, Electroporation methods, RNA, Messenger immunology
- Abstract
The immune system is a crucial player in the development of cancer. Once it is in imbalance and immunosuppressive mechanisms supporting tumor growth take over control, dendritic cell immunotherapy might offer a solution to restore the balance. There are several methods to manufacture dendritic cells but none of them has yet proven to be superior to others. In this chapter, we discuss the methodology using electroporation of mRNA encoding Wilms' tumor gene 1, survivin, and TriMix (mixture of caTLR4, CD40L, and CD70) to simultaneously load and mature dendritic cells.
- Published
- 2016
- Full Text
- View/download PDF
35. Long-term clinical outcome of melanoma patients treated with messenger RNA-electroporated dendritic cell therapy following complete resection of metastases.
- Author
-
Wilgenhof S, Corthals J, Van Nuffel AM, Benteyn D, Heirman C, Bonehill A, Thielemans K, and Neyns B
- Subjects
- Adult, Aged, Cancer Vaccines genetics, Cancer Vaccines immunology, Electroporation, Female, Humans, Interferon alpha-2, Interferon-alpha administration & dosage, Interferon-alpha immunology, MART-1 Antigen genetics, Male, Melanoma immunology, Melanoma surgery, Melanoma-Specific Antigens genetics, Middle Aged, Monophenol Monooxygenase genetics, Neoplasm Metastasis, Pilot Projects, RNA, Messenger genetics, Recombinant Proteins administration & dosage, Recombinant Proteins immunology, Skin Neoplasms immunology, Skin Neoplasms surgery, Treatment Outcome, Melanoma, Cutaneous Malignant, Cancer Vaccines administration & dosage, Dendritic Cells immunology, Immunotherapy, Adoptive methods, Melanoma therapy, RNA, Messenger administration & dosage, Skin Neoplasms therapy
- Abstract
Purpose: Melanoma patients with a high risk of recurrence may benefit from immunotherapy with mRNA-electroporated autologous monocyte-derived dendritic cells (DCs). Further benefit may be found in combining DC-therapy with interferon alfa-2b., Patients and Methods: The long-term clinical outcome of AJCC stage III/IV melanoma patients who had no evidence of disease at the time of treatment with autologous mRNA-electroporated DCs in a single-center pilot clinical trial was analyzed. Antigen loading was accomplished by co-electroporation of mRNA encoding a fusion protein between MAGE-A1, -A3, -C2, Tyrosinase, MelanA/MART-1, or gp100, and an HLA class II-targeting sequence. DCs were administered by 4-6 bi-weekly intradermal injections. IFN-α-2b (5 MIU TIW) was initiated either at recurrence (cohort 1), concomitant with DCs (cohorts 2 and 3), or following the fourth DC administration (cohort 4)., Results: Thirty melanoma patients were recruited between April 2006 and June 2009. DC-related adverse events included grade 2 local injection site reactions in all patients, grade 2 fever and flu-like symptoms in one patient, and skin depigmentation in seven patients. After a median follow-up of over 6 years, the median relapse-free survival is 22 months (95% CI 12-32 months). Twelve patients have died. The median overall survival has not been reached; the 2-year and 4-year survival rates are 93 and 70%, respectively., Conclusions: Adjuvant therapy following the resection of melanoma metastases with autologous mRNA-electroporated DCs, combined with interferon alfa-2b, is tolerable and results in encouraging long-term overall survival rates justifying further evaluation in a randomized clinical trial.
- Published
- 2015
- Full Text
- View/download PDF
36. Immunomodulatory drugs improve the immune environment for dendritic cell-based immunotherapy in multiple myeloma patients after autologous stem cell transplantation.
- Author
-
De Keersmaecker B, Fostier K, Corthals J, Wilgenhof S, Heirman C, Aerts JL, Thielemans K, and Schots R
- Subjects
- Adult, Angiogenesis Inhibitors therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cell Proliferation, Dexamethasone administration & dosage, Doxorubicin administration & dosage, Female, Humans, Immunomodulation, Lenalidomide, Male, Melphalan administration & dosage, Middle Aged, Multiple Myeloma drug therapy, Multiple Myeloma immunology, Thalidomide therapeutic use, Transplantation Conditioning methods, Transplantation, Autologous, Vincristine administration & dosage, Dendritic Cells immunology, Hematopoietic Stem Cell Transplantation methods, Immunologic Factors therapeutic use, Immunotherapy, Adoptive methods, Multiple Myeloma therapy, Thalidomide analogs & derivatives
- 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
37. Optimized dendritic cell-based immunotherapy for melanoma: the TriMix-formula.
- Author
-
Van Lint S, Wilgenhof S, Heirman C, Corthals J, Breckpot K, Bonehill A, Neyns B, and Thielemans K
- Subjects
- Animals, Humans, Dendritic Cells immunology, Immunotherapy, Adoptive methods, Melanoma immunology, Melanoma therapy
- Abstract
Since decades, the main goal of tumor immunologists has been to increase the capacity of the immune system to mediate tumor regression. In this regard, one of the major focuses of cancer immunotherapy has been the design of vaccines promoting strong tumor-specific cytotoxic T lymphocyte responses in cancer patients. Here, dendritic cells (DCs) play a pivotal role as they are regarded as nature's adjuvant and as such have become the natural agents for antigen delivery in order to finally elicit strong T cell responses (Villadangos and Schnorrer in Nat Rev Immunol 7:543-555, 2007; Melief in Immunity 29:372-383, 2008; Palucka and Banchereau in Nat Rev Cancer 12:265-277, 2012; Vacchelli et al. in Oncoimmunology 2:e25771, 2013; Galluzzi et al. in Oncoimmunology 1:1111-1134, 2012). Therefore, many investigators are actively pursuing the use of DCs as an efficient way of inducing anticancer immune responses. Nowadays, DCs can be generated at a large scale in closed systems, yielding sufficient numbers of cells for clinical application. In addition, with the identification of tumor-associated antigens, which are either selectively or preferentially expressed by tumors, a whole range of strategies using DCs for immunotherapy have been designed and tested in clinical studies. Despite the evidence that DCs loaded with tumor-associated antigens can elicit immune responses in vivo, clinical responses remained disappointingly low. Therefore, optimization of the cellular product and route of administration was urgently needed. Here, we review the path we have followed in the development of TriMixDC-MEL, a potent DC-based cellular therapy, discussing its development as well as further modifications and applications.
- Published
- 2014
- Full Text
- View/download PDF
38. AZD1480 delays tumor growth in a melanoma model while enhancing the suppressive activity of myeloid-derived suppressor cells.
- Author
-
Maenhout SK, Du Four S, Corthals J, Neyns B, Thielemans K, and Aerts JL
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Disease Models, Animal, Female, Humans, Janus Kinase 2 antagonists & inhibitors, Melanoma immunology, Melanoma pathology, Melanoma, Experimental drug therapy, Melanoma, Experimental immunology, Melanoma, Experimental pathology, Mice, Mice, Inbred C57BL, Myeloid Cells immunology, Random Allocation, Signal Transduction, Xenograft Model Antitumor Assays, Melanoma drug therapy, Myeloid Cells drug effects, Pyrazoles pharmacology, Pyrimidines pharmacology
- 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
39. Design of an Optimized Wilms' Tumor 1 (WT1) mRNA Construct for Enhanced WT1 Expression and Improved Immunogenicity In Vitro and In Vivo.
- Author
-
Benteyn D, Anguille S, Van Lint S, Heirman C, Van Nuffel AM, Corthals J, Ochsenreither S, Waelput W, Van Beneden K, Breckpot K, Van Tendeloo V, Thielemans K, and Bonehill A
- Abstract
Tumor antigen-encoding mRNA for dendritic cell (DC)-based vaccination has gained increasing popularity in recent years. Within this context, two main strategies have entered the clinical trial stage: the use of mRNA for ex vivo antigen loading of DCs and the direct application of mRNA as a source of antigen for DCs in vivo. DCs transfected with mRNA-encoding Wilms' tumor 1 (WT1) protein have shown promising clinical results. Using a stepwise approach, we re-engineered a WT1 cDNA-carrying transcription vector to improve the translational characteristics and immunogenicity of the transcribed mRNA. Different modifications were performed: (i) the WT1 sequence was flanked by the lysosomal targeting sequence of dendritic cell lysosomal-associated membrane protein to enhance cytoplasmic expression; (ii) the nuclear localization sequence (NLS) of WT1 was deleted to promote shuttling from the nucleus to the cytoplasm; (iii) the WT1 DNA sequence was optimized in silico to improve translational efficiency; and (iv) this WT1 sequence was cloned into an optimized RNA transcription vector. DCs electroporated with this optimized mRNA showed an improved ability to stimulate WT1-specific T-cell immunity. Furthermore, in a murine model, we were able to show the safety, immunogenicity, and therapeutic activity of this optimized mRNA. This work is relevant for the future development of improved mRNA-based vaccine strategies K.Molecular Therapy-Nucleic Acids (2013) 2, e134; doi:10.1038/mtna.2013.54; published online 19 November 2013.
- Published
- 2013
- Full Text
- View/download PDF
40. Modulation of regulatory T cell function by monocyte-derived dendritic cells matured through electroporation with mRNA encoding CD40 ligand, constitutively active TLR4, and CD70.
- Author
-
Pen JJ, De Keersmaecker B, Maenhout SK, Van Nuffel AM, Heirman C, Corthals J, Escors D, Bonehill A, Thielemans K, Breckpot K, and Aerts JL
- Subjects
- CD27 Ligand genetics, CD4-Positive T-Lymphocytes cytology, CD40 Ligand genetics, Cell Differentiation drug effects, Cell Division, Cells, Cultured, Coculture Techniques, Cytokines pharmacology, Dendritic Cells metabolism, Electroporation, Humans, Immunophenotyping, Lymphocyte Activation, MAP Kinase Kinase 1 genetics, MAP Kinase Kinase 1 physiology, Monocytes cytology, Monocytes drug effects, RNA, Messenger administration & dosage, RNA, Messenger genetics, Recombinant Fusion Proteins metabolism, T-Lymphocytes, Regulatory cytology, Th1 Cells immunology, Toll-Like Receptor 4 genetics, CD27 Ligand physiology, CD40 Ligand physiology, CD8-Positive T-Lymphocytes immunology, Dendritic Cells immunology, Immune Tolerance immunology, Lymphopoiesis physiology, T-Lymphocytes, Regulatory immunology, Toll-Like Receptor 4 physiology
- Abstract
Regulatory T cells (Tregs) counteract anticancer immune responses through a number of mechanisms, limiting dendritic cell (DC)-based anticancer immunotherapy. In this study, we investigated the influence of various DC activation stimuli on the Treg functionality. We compared DCs activated by electroporation with mRNA encoding constitutively active TLR4 (caTLR4) and CD40 ligand (DiMix-DCs), or these factors together with mRNA encoding the costimulatory molecule CD70 (TriMix-DCs) with DCs maturated in the presence of a mixture of inflammatory cytokines (DCs maturated with a combination of the cytokines IL-1β, IL-6, TNF-α, and PGE2) for their ability to counteract Tregs on different levels. We first demonstrated that there was no difference in the extent of Treg induction starting from CD4(+)CD25(-) T cells under the influence of the different DC maturation stimuli. Second, we showed that both DiMix- and TriMix-DCs could partly alleviate Treg inhibition of CD8(+) T cells. Third, we observed that CD8(+) T cells that had been precultured with DiMix-DCs or TriMix-DCs were partially protected against subsequent Treg suppression. Finally, we showed that Tregs cocultured in the presence of TriMix-DCs, but not DiMix-DCs, partially lost their suppressive capacity. This was accompanied by a decrease in CD27 and CD25 expression on Tregs, as well as an increase in the expression of T-bet and secretion of IFN-γ, TNF-α, and IL-10, suggesting a shift of the Treg phenotype toward a Th1 phenotype. In conclusion, these data suggest that TriMix-DCs are not only able to suppress Treg functions, but moreover could be able to reprogram Tregs to Th1 cells under certain circumstances.
- Published
- 2013
- Full Text
- View/download PDF
41. Characterization of CD8+ T-cell responses in the peripheral blood and skin injection sites of melanoma patients treated with mRNA electroporated autologous dendritic cells (TriMixDC-MEL).
- Author
-
Benteyn D, Van Nuffel AM, Wilgenhof S, Corthals J, Heirman C, Neyns B, Thielemans K, and Bonehill A
- Subjects
- Biopsy, Cell Movement, Electroporation, Epitopes immunology, Humans, Neoplasm Metastasis, Pilot Projects, Recurrence, Skin metabolism, Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, Cancer Vaccines therapeutic use, Dendritic Cells transplantation, Melanoma therapy, Skin Neoplasms therapy
- Abstract
Treatment of melanoma patients with mRNA electroporated dendritic cells (TriMixDC-MEL) stimulates T-cell responses against the presented tumor-associated antigens (TAAs). In the current clinical trials, melanoma patients with systemic metastases are treated, requiring priming and/or expansion of preexisting TAA-specific T cells that are able to migrate to both the skin and internal organs. We monitored the presence of TAA-specific CD8(+) T cells infiltrating the skin at sites of intradermal TriMixDC-MEL injection (SKILs) and within the circulation of melanoma patients treated in two clinical trials. In 10 out of fourteen (71%) patients screened, CD8(+) T cells recognizing any of the four TAA presented by TriMixDC-MEL cellular vaccine were found in both compartments. In total, 30 TAA-specific T-cell responses were detected among the SKILs and 29 among peripheral blood T cells, of which 24 in common. A detailed characterization of the antigen specificity of CD8(+) T-cell populations in four patients indicates that the majority of the epitopes detected were only recognized by CD8(+) T cells derived from either skin biopsies or peripheral blood, indicating that some compartmentalization occurs after TriMix-DC therapy. To conclude, functional TAA-specific CD8(+) T cells distribute both to the skin and peripheral blood of patients after TriMixDC-MEL therapy.
- Published
- 2013
- Full Text
- View/download PDF
42. Intravenous and intradermal TriMix-dendritic cell therapy results in a broad T-cell response and durable tumor response in a chemorefractory stage IV-M1c melanoma patient.
- Author
-
Van Nuffel AM, Benteyn D, Wilgenhof S, Corthals J, Heirman C, Neyns B, Thielemans K, and Bonehill A
- Subjects
- Amino Acid Sequence, CD27 Ligand biosynthesis, CD27 Ligand genetics, CD27 Ligand immunology, CD40 Ligand biosynthesis, CD40 Ligand genetics, CD40 Ligand immunology, Dendritic Cells pathology, Electroporation methods, Humans, Hypersensitivity, Delayed immunology, Male, Melanoma pathology, Middle Aged, Molecular Sequence Data, Neoplasm Staging, Prospective Studies, RNA, Messenger administration & dosage, RNA, Messenger genetics, Toll-Like Receptor 4 biosynthesis, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 immunology, Dendritic Cells immunology, Helium administration & dosage, Immunotherapy, Adoptive methods, Melanoma immunology, Melanoma therapy, Nitrogen administration & dosage, Oxygen administration & dosage, T-Lymphocytes immunology
- Abstract
Dendritic cells (DCs) electroporated with mRNA encoding CD70, CD40L and a constitutively active toll-like receptor 4 (TriMix-DC) have an increased T-cell stimulatory capacity. In a prospective phase IB clinical trial, we treated melanoma patients with intradermal and intravenous injections of autologous TriMix-DC co-electroporated with mRNA encoding full-length MAGE-A3, MAGE-C2, tyrosinase and gp100. We report here the immunological and clinical results obtained in one patient with a particularly favorable outcome. This patient had stage IV-M1c melanoma with documented progression during dacarbazine chemotherapy and received 5 TriMix-DC injections. Following DC therapy, a broad CD8(+) T-cell response against multiple epitopes derived from all four treatment antigens was found in the blood and among T cells derived from DTH biopsy. In addition, CD4(+) T cells recognizing different MAGE-A3-derived epitopes were detected in DTH-derived cells. A spontaneous anti-MAGE-C2 CD8(+) T-cell response was present prior to TriMix-DC therapy and increased during treatment. The tumor response was assessed with 18-fluorodeoxyglucose-positron emission/computed tomography. We documented a partial tumor response according to RECIST criteria with a marked reduction in (18)F-FDG-uptake by lung, lymph node and bone metastases. The patient remains free from progression after 12 months of follow-up. This case report indicates that administration of autologous TriMix-DC by the combined intradermal and intravenous route can mediate a durable objective tumor response accompanied by a broad T-cell response in a chemorefractory stage IV-M1c melanoma patient.
- Published
- 2012
- Full Text
- View/download PDF
43. Dendritic cells loaded with mRNA encoding full-length tumor antigens prime CD4+ and CD8+ T cells in melanoma patients.
- Author
-
Van Nuffel AM, Benteyn D, Wilgenhof S, Pierret L, Corthals J, Heirman C, van der Bruggen P, Coulie PG, Neyns B, Thielemans K, and Bonehill A
- Subjects
- Amino Acid Sequence, Antigen Presentation, Antigens, Neoplasm genetics, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Cytotoxicity, Immunologic, Dendritic Cells immunology, Electroporation, HLA-D Antigens genetics, Histocompatibility Antigens Class I genetics, Humans, Lymphocyte Activation, Melanoma immunology, Melanoma pathology, Molecular Sequence Data, Protein Sorting Signals, RNA, Messenger genetics, Skin Neoplasms immunology, Skin Neoplasms pathology, Transfection, Antigens, Neoplasm immunology, Dendritic Cells transplantation, HLA-D Antigens immunology, Histocompatibility Antigens Class I immunology, Immunotherapy, Adoptive methods, Melanoma therapy, RNA, Messenger immunology, Skin Neoplasms therapy
- Abstract
It is generally thought that dendritic cells (DCs) loaded with full-length tumor antigen could improve immunotherapy by stimulating broad T-cell responses and by allowing treatment irrespective of the patient's human leukocyte antigen (HLA) type. To investigate this, we determined the specificity of T cells from melanoma patients treated with DCs loaded with mRNA encoding a full-length tumor antigen fused to a signal peptide and an HLA class II sorting signal, allowing presentation in HLA class I and II. In delayed-type hypersensitive (DTH)-biopsies and blood, we found functional CD8(+) and CD4(+) T cells recognizing novel treatment-antigen-derived epitopes, presented by several HLA types. Additionally, we identified a CD8(+) response specific for the signal peptide incorporated to elicit presentation by HLA class II and a CD4(+) response specific for the fusion region of the signal peptide and one of the antigens. This demonstrates that the fusion proteins contain newly created immunogenic sequences and provides evidence that ex vivo-generated mRNA-modified DCs can induce effector CD8(+) and CD4(+) T cells from the naive T-cell repertoire of melanoma patients. Thus, this work provides definitive proof that DCs presenting the full antigenic spectrum of tumor antigens can induce T cells specific for novel epitopes and can be administered to patients irrespective of their HLA type.
- Published
- 2012
- Full Text
- View/download PDF
44. Epitope and HLA-type independent monitoring of antigen-specific T-cells after treatment with dendritic cells presenting full-length tumor antigens.
- Author
-
Van Nuffel AM, Tuyaerts S, Benteyn D, Wilgenhof S, Corthals J, Heirman C, Neyns B, Thielemans K, and Bonehill A
- Subjects
- Antigens, Neoplasm immunology, Biopsy, Flow Cytometry, Humans, Hypersensitivity, Delayed immunology, Immunophenotyping, Interferon-gamma analysis, Interferon-gamma immunology, Lysosomal-Associated Membrane Protein 1 analysis, Lysosomal-Associated Membrane Protein 1 immunology, Skin cytology, Skin immunology, Tumor Necrosis Factor Receptor Superfamily, Member 9 analysis, Tumor Necrosis Factor Receptor Superfamily, Member 9 immunology, Tumor Necrosis Factor-alpha analysis, Tumor Necrosis Factor-alpha immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Dendritic Cells immunology, Epitopes, T-Lymphocyte immunology, Immunotherapy, Adoptive methods, Melanoma immunology, Melanoma therapy
- Abstract
The efficacy of cancer immunotherapy can be improved by treatment with full-length tumor antigen and by combining several antigens. This approach allows the induction of a broad immune response irrespective of the patient's HLA type which at the same time challenges immune monitoring. Also, the number of available lymphocytes is most often limited and minimal in vitro restimulations of the lymphocytes should maintain information about the actual in vivo situation. To overcome these hurdles, we developed a method to measure the CD8(+) and CD4(+) T-cell responses directly ex vivo. Skin biopsies taken from dendritic cell (DC)-induced DTH reactions from melanoma patients participating in a DC-clinical trial served as lymphocyte source. Antigen-specificity of skin infiltrating lymphocytes was investigated by coculture with antigen-presenting autologous B cells and assessed for CD137 upregulation and enhanced cytokine secretion. Using this approach we could detect treatment-specific CD8(+) T-cells without restimulation in vitro. Upregulation of the activation marker CD137 correlated with the upregulation of the lytic marker CD107a. CD137 upregulation by treatment-specific CD4(+) lymphocytes however was more pronounced after antigen-specific in vitro restimulation. Both CD8(+) and CD4(+) lymphocytes could be further expanded using the same B cells as for screening allowing characterization of the recognized antigenic region. In addition, this technique can be extended to detect a broader array of T-cell functions and to monitor a large cohort of patients. We believe that this approach of direct ex vivo monitoring, irrespective of the patient's HLA-type or the recognized peptide, and using a limited number of lymphocytes is a valuable tool in the immune monitoring of current cellular immunotherapies., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
45. A phase I/IIa immunotherapy trial of HIV-1-infected patients with Tat, Rev and Nef expressing dendritic cells followed by treatment interruption.
- Author
-
Allard SD, De Keersmaecker B, de Goede AL, Verschuren EJ, Koetsveld J, Reedijk ML, Wylock C, De Bel AV, Vandeloo J, Pistoor F, Heirman C, Beyer WE, Eilers PH, Corthals J, Padmos I, Thielemans K, Osterhaus AD, Lacor P, van der Ende ME, Aerts JL, van Baalen CA, and Gruters RA
- Subjects
- Adult, Aged, Cells, Cultured, Gene Products, rev immunology, Gene Products, tat immunology, HIV Infections immunology, Humans, Male, Middle Aged, nef Gene Products, Human Immunodeficiency Virus immunology, AIDS Vaccines immunology, Dendritic Cells immunology, HIV Infections therapy, HIV-1 immunology, Immunization
- Abstract
In a phase I/IIa clinical trial, 17 HIV-1 infected patients, stable on cART, received 4 vaccinations with autologous dendritic cells electroporated with mRNA encoding Tat, Rev and Nef, after which cART was interrupted. Vaccination was safe and feasible. During the analytical treatment interruption (ATI), no serious adverse events were observed. Ninety-six weeks following ATI, 6/17 patients remained off therapy. Although induced and/or enhanced CD4(+) and CD8(+) T-cell responses specific for the immunogens were observed in most of the patients, we found no correlation with the number of weeks off cART. Moreover, CD4(+) T-cell counts, plasma viral load and the time remaining off cART following ATI did not differ from historical control data. To conclude, the vaccine was safe, well tolerated and resulted in vaccine-specific immune responses. Since no correlation with clinical parameters could be found, these results warrant further research in order to optimize the efficacy of vaccine-induced T-cell responses., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
46. Therapeutic vaccination with an autologous mRNA electroporated dendritic cell vaccine in patients with advanced melanoma.
- Author
-
Wilgenhof S, Van Nuffel AM, Corthals J, Heirman C, Tuyaerts S, Benteyn D, De Coninck A, Van Riet I, Verfaillie G, Vandeloo J, Bonehill A, Thielemans K, and Neyns B
- Subjects
- Adult, Aged, CD27 Ligand immunology, CD27 Ligand metabolism, CD40 Antigens immunology, CD40 Antigens metabolism, Cancer Vaccines administration & dosage, Dendritic Cells cytology, Dendritic Cells metabolism, Electroporation, Female, Histocompatibility Antigens Class II immunology, Histocompatibility Antigens Class II metabolism, Humans, Hypersensitivity, Delayed immunology, Interferon alpha-2, Male, Middle Aged, Neoplasm Staging, RNA, Messenger metabolism, Recombinant Proteins, Skin Neoplasms immunology, Skin Neoplasms mortality, Skin Neoplasms pathology, Survival Analysis, Toll-Like Receptor 4 immunology, Toll-Like Receptor 4 metabolism, Vaccination, Antigens, Neoplasm immunology, Cancer Vaccines immunology, Dendritic Cells immunology, Drug Therapy, Combination methods, Interferon-alpha administration & dosage, Melanoma, RNA, Messenger immunology, Skin Neoplasms drug therapy
- Abstract
The immunostimulatory capacity of dendritic cells is improved by co-electroporation with mRNA encoding CD40 ligand, constitutively active toll-like receptor 4, and CD70 (TriMix-DC). This pilot clinical trial evaluated the feasibility, safety, and immunogenicity of a therapeutic vaccination containing autologous TriMix-DC co-electroporated with mRNA encoding a human leukocyte antigen class II-targeting signal linked to 1 of 4 melanoma-associated antigens (MAGE-A3, MAGE-C2, tyrosinase, and gp100) in patients with advanced melanoma. Thirty-five American Joint Committee on Cancer stage III/IV melanoma patients received autologous TriMix-DC (4 administrations 2 weeks apart). Immune monitoring was performed by evaluating skin biopsies of delayed type IV hypersensitivity (DTH) reactions for presence of vaccinal antigen-specific DTH-infiltrating lymphocytes (DIL). Thereafter, patients could receive interferon-alpha-2b (IFN-α-2b) 5 MU subcutaneously 3 times weekly and additional TriMix-DC every 8 weeks. TriMix-DC-related adverse events comprised grade 2 local injection site reactions (all patients), and grade 2 fever and lethargy (2 patients). Vaccinal antigen-specific DIL were found in 0/6 patients tested at vaccine initiation and in 12/21 (57.1%) assessed after the fourth vaccine. A positive postvaccination DTH test correlated with IL-12p70 secretion capacity of TriMix-DC. No objective responses to TriMix-DC alone were seen according to RECIST. Twenty-nine patients received IFN-α-2b after the fourth vaccine without unexpected adverse events. During TriMix-DC/IFN-α-2b combination therapy, 1 partial response and 5 stable disease (disease control of >6 months with regression of metastases) were observed in 17 patients with evaluable disease at baseline. In conclusion, this study demonstrated that therapeutic vaccination with autologous TriMix-DC is feasible, safe, and immunogenic and can be combined with sequential IFN-α-2b.
- Published
- 2011
- Full Text
- View/download PDF
47. The combination of 4-1BBL and CD40L strongly enhances the capacity of dendritic cells to stimulate HIV-specific T cell responses.
- Author
-
De Keersmaecker B, Heirman C, Corthals J, Empsen C, van Grunsven LA, Allard SD, Pen J, Lacor P, Thielemans K, and Aerts JL
- Subjects
- Blotting, Western, CD40 Antigens immunology, Cell Proliferation, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, HIV Seropositivity, Humans, Lymphocyte Activation, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, T-Lymphocytes, Regulatory immunology, 4-1BB Ligand pharmacology, CD40 Ligand pharmacology, Dendritic Cells immunology, HIV immunology, HIV Infections immunology, T-Lymphocytes immunology
- Abstract
One of the consequences of HIV infection is a progressive loss of T cell functions, resulting in decreased cytokine secretion and proliferation and an increased sensitivity to apoptosis. Therefore, successful therapeutic vaccination approaches should aim at restoring the functionality of existing HIV-specific T cells, as well as to efficiently induce potent, HIV-specific T cells from naïve T cells. In this study, we wanted to determine the stimulatory capacity of DCs coelectroporated with mRNA encoding for different costimulatory molecules of the TNFSF, together with HIV antigen-encoding mRNA. We show that DCs electroporated with 4-1BBL can enhance the proliferation, functionality, cytokine production, and survival of HIV-specific CD8(+) T cells. Furthermore, we are the first to show that a combination of 4-1BBL and CD40L overexpression on DCs dramatically enhances CD4(+) and CD8(+) T cell responses. Finally, we demonstrate that signaling through 4-1BB, but not through CD40, can alleviate the suppressive effect of Tregs on CD8(+) T cell proliferation. Thus, the combination of 4-1BBL and CD40L enhances HIV-specific CD8(+) T cell responses in a synergistic way, resulting in enhanced proliferation of CD4(+) and CD8(+) T cell subsets, an increased cytokine secretion, and a reduced sensitivity to Treg-mediated immune suppression.
- Published
- 2011
- Full Text
- View/download PDF
48. Restoration of tumor equilibrium after immunotherapy for advanced melanoma: three illustrative cases.
- Author
-
Wilgenhof S, Pierret L, Corthals J, Van Nuffel AM, Heirman C, Roelandt T, De Coninck A, Verfaillie G, Vandenbroucke F, Van Riet I, Bonehill A, Thielemans K, and Neyns B
- Subjects
- Adult, Aged, Female, Humans, Ipilimumab, Male, Melanoma pathology, Melanoma secondary, Middle Aged, Neoplasm Metastasis, Antibodies, Monoclonal therapeutic use, Immunotherapy methods, Melanoma immunology, Melanoma therapy
- Abstract
Metastatic melanoma runs a predictable detrimental course in the vast majority of patients. New modalities of immunotherapy, such as melanoma antigen-specific therapeutic vaccination and cytotoxic T-lymphocyte antigen 4 (CTLA-4) receptor blockade by monoclonal antibodies (mAbs), have been associated with atypical kinetics of tumor response that differ from those observed during cytotoxic treatment. Recently, new tumor response criteria have been proposed based on the tumor response characteristics observed in clinical studies with ipilimumab (the so-called 'immune-related response criteria'). We report three illustrative cases of the American Joint Committee on Cancer stage IV-M1c melanoma patients who experienced atypical kinetics of tumor response to the treatment with the CTLA-4-blocking mAb, ipilimumab (case 1), or an autologous dendritic cell vaccine in combination with interferon α-2b (cases 2 and 3). These cases show that atypical response patterns not only relate to the outcome of CTLA-4-blocking mAb therapy but also to the treatment with therapeutic vaccines and interferon α-2b.
- Published
- 2011
- Full Text
- View/download PDF
49. Lumenal part of the DC-LAMP protein is not required for induction of antigen-specific T cell responses by means of antigen-DC-LAMP messenger RNA-electroporated dendritic cells.
- Author
-
De Keersmaecker B, Heirman C, Allard S, Bonehill A, Corthals J, Thielemans K, and Aerts JL
- Subjects
- Antigen Presentation, Cells, Cultured, Dendritic Cells metabolism, Gene Products, gag genetics, Gene Products, gag metabolism, HIV Infections immunology, HIV Infections virology, HIV-1 immunology, Humans, Lymphocyte Activation, RNA, Messenger genetics, Dendritic Cells immunology, Electroporation, Gene Products, gag immunology, Lysosomal Membrane Proteins chemistry, Lysosomal Membrane Proteins genetics, Lysosomal Membrane Proteins immunology, Lysosomal Membrane Proteins metabolism, RNA, Messenger metabolism, T-Lymphocytes immunology
- Abstract
Previous studies showed that stimulation of T cells derived from HIV-1-infected patients with autologous dendritic cells electroporated with mRNA encoding HIV antigens can induce antigen-specific T cell responses in vitro. Linking the antigen to an MHC class II-targeting sequence, such as dendritic cell lysosome-associated membrane protein (DC-LAMP), in the mRNA construct results in presentation of antigenic peptides in both MHC class I and class II molecules and therefore enhances the induced T cell responses. To analyze whether the lumenal domain of DC-LAMP is required for optimal induction of cellular immunity against HIV antigens, we compared fusion constructs with or without the lumenal domain of the DC-LAMP protein. A human codon-optimized consensus Gag sequence and a chimeric cDNA sequence encompassing Tat, Rev, and Nef codons (TaReNef ) were cloned into a vector containing the DC-LAMP sequence with or without its lumenal domain. The Gag protein lacking the DC-LAMP-derived sequence altogether elicited only weak T cell responses. DCs electroporated with Gag or TaReNef linked to DC-LAMP were able to elicit similar levels of antigen-specific CD4(+) and CD8(+) T cell responses for both Gag and TaReNef, irrespective of the addition of the DC-LAMP lumenal domain. These data show that DC-LAMP-mediated antigen targeting is absolutely required for optimal T cell stimulation, but that in our experimental setup, the lumenal part of DC-LAMP does not improve the overall induction of antigen-specific T cell responses.
- Published
- 2010
- Full Text
- View/download PDF
50. Immunotherapy of cancer with dendritic cells loaded with tumor antigens and activated through mRNA electroporation.
- Author
-
Van Nuffel AM, Corthals J, Neyns B, Heirman C, Thielemans K, and Bonehill A
- Subjects
- Cancer Vaccines administration & dosage, Cancer Vaccines immunology, Cell Differentiation, Dendritic Cells cytology, Humans, Monitoring, Immunologic, RNA Caps biosynthesis, RNA, Messenger genetics, RNA, Messenger metabolism, Vaccination, Antigens, Neoplasm immunology, Dendritic Cells immunology, Electroporation methods, Immunotherapy methods, Melanoma immunology, Melanoma therapy
- Abstract
Since decades, the main goal of tumor immunologists has been to increase the capacity of the immune system to mediate tumor regression. Considerable progress has been made in enhancing the efficacy of therapeutic anticancer vaccines. First, dendritic cells (DCs) have been identified as the key players in orchestrating primary immune responses. A better understanding of their biology and the development of procedures to generate vast amounts of DCs in vitro have accelerated the development of potent immunotherapeutic strategies for cancer. Second, tumor-associated antigens have been identified which are either selectively or preferentially expressed by tumor cells and can be recognized by the immune system. Finally, several studies have been performed on the genetic modification of DCs with tumor antigens. In this regard, loading the DCs with mRNA, which enables them to produce/process and present the tumor antigens themselves, has emerged as a promising strategy. Here, we will first overview the different aspects that must be taken into account when generating an mRNA-based DC vaccine and the published clinical studies exploiting mRNA-loaded DCs. Second, we will give a detailed description of a novel procedure to generate a vaccine consisting of tumor antigen-expressing dendritic cells with an in vitro superior capacity to induce anti-tumor immune responses. Here, immature DCs are electroporated with mRNAs encoding a tumor antigen, CD40 ligand (CD40L), CD70, and constitutively active (caTLR4) to generate mature antigen-presenting DCs.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.