9 results on '"de Goede AL"'
Search Results
2. 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
3. Adjuvant dendritic cell therapy in stage IIIB/C melanoma: the MIND-DC randomized phase III trial.
- Author
-
Bol KF, Schreibelt G, Bloemendal M, van Willigen WW, Hins-de Bree S, de Goede AL, de Boer AJ, Bos KJH, Duiveman-de Boer T, Olde Nordkamp MAM, van Oorschot TGM, Popelier CJ, Pots JM, Scharenborg NM, van de Rakt MWMM, de Ruiter V, van Meeteren WS, van Rossum MM, Croockewit SJ, Koeneman BJ, Creemers JHA, Wortel IMN, Angerer C, Brüning M, Petry K, Dzionek A, van der Veldt AA, van Grünhagen DJ, Werner JEM, Bonenkamp JJ, Haanen JBAG, Boers-Sonderen MJ, Koornstra RHT, Boomsma MF, Aarntzen EHJ, Gotthardt M, Nagarajah J, de Witte TJM, Figdor CG, de Wilt JHW, Textor J, de Groot JWB, Gerritsen WR, and de Vries IJM
- Subjects
- Humans, Disease-Free Survival, Adjuvants, Immunologic therapeutic use, Dendritic Cells pathology, Neoplasm Staging, Melanoma, Skin Neoplasms pathology
- Abstract
Autologous natural dendritic cells (nDCs) treatment can induce tumor-specific immune responses and clinical responses in cancer patients. In this phase III clinical trial (NCT02993315), 148 patients with resected stage IIIB/C melanoma were randomized to adjuvant treatment with nDCs (n = 99) or placebo (n = 49). Active treatment consisted of intranodally injected autologous CD1c+ conventional and plasmacytoid DCs loaded with tumor antigens. The primary endpoint was the 2-year recurrence-free survival (RFS) rate, whereas the secondary endpoints included median RFS, 2-year and median overall survival, adverse event profile, and immunological response The 2-year RFS rate was 36.8% in the nDC treatment group and 46.9% in the control group (p = 0.31). Median RFS was 12.7 months vs 19.9 months, respectively (hazard ratio 1.25; 90% CI: 0.88-1.79; p = 0.29). Median overall survival was not reached in both treatment groups (hazard ratio 1.32; 90% CI: 0.73-2.38; p = 0.44). Grade 3-4 study-related adverse events occurred in 5% and 6% of patients. Functional antigen-specific T cell responses could be detected in 67.1% of patients tested in the nDC treatment group vs 3.8% of patients tested in the control group (p < 0.001). In conclusion, while adjuvant nDC treatment in stage IIIB/C melanoma patients generated specific immune responses and was well tolerated, no benefit in RFS was observed., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
4. Good manufacturing practice production of CD34 + progenitor-derived NK cells for adoptive immunotherapy in acute myeloid leukemia.
- Author
-
de Jonge PKJD, van Hauten PMM, Janssen LD, de Goede AL, Berrien-Elliott MM, van der Meer JMR, Mousset CM, Roeven MWH, Foster M, Blijlevens N, Hobo W, Fehniger TA, Jansen JH, Schaap NPM, and Dolstra H
- Subjects
- Humans, Killer Cells, Natural metabolism, Antigens, CD34 metabolism, Hematopoietic Stem Cells, Immunotherapy, Adoptive methods, Leukemia, Myeloid, Acute genetics
- Abstract
Allogeneic natural killer (NK) cell-based immunotherapy is a promising, well-tolerated adjuvant therapeutic approach for acute myeloid leukemia (AML). For reproducible NK cell immunotherapy, a homogenous, pure and scalable NK cell product is preferred. Therefore, we developed a good manufacturing practice (GMP)-compliant, cytokine-based ex vivo manufacturing process for generating NK cells from CD34
+ hematopoietic stem and progenitor cells (HSPC). This manufacturing process combines amongst others IL15 and IL12 and the aryl hydrocarbon receptor antagonist StemRegenin-1 (SR1) to generate a consistent and active NK cell product that fits the requirements for NK cell immunotherapy well. The cell culture protocol was first optimized to generate NK cells with required expansion and differentiation capacity in GMP-compliant closed system cell culture bags. In addition, phenotype, antitumor potency, proliferative and metabolic capacity were evaluated to characterize the HSPC-NK product. Subsequently, seven batches were manufactured for qualification of the process. All seven runs demonstrated consistent results for proliferation, differentiation and antitumor potency, and preliminary specifications for the investigational medicinal product for early clinical phase trials were set. This GMP-compliant manufacturing process for HSPC-NK cells (named RNK001 cells) is used to produce NK cell batches applied in the clinical trial 'Infusion of ex vivo-generated allogeneic natural killer cells in combination with subcutaneous IL2 in patients with acute myeloid leukemia' approved by the Dutch Ethics Committee (EudraCT 2019-001929-27)., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
5. Convection Enhanced Delivery of the Oncolytic Adenovirus Delta24-RGD in Patients with Recurrent GBM: A Phase I Clinical Trial Including Correlative Studies.
- Author
-
van Putten EHP, Kleijn A, van Beusechem VW, Noske D, Lamers CHJ, de Goede AL, Idema S, Hoefnagel D, Kloezeman JJ, Fueyo J, Lang FF, Teunissen CE, Vernhout RM, Bakker C, Gerritsen W, Curiel DT, Vulto A, Lamfers MLM, and Dirven CMF
- Subjects
- Adenoviridae genetics, Convection, Humans, Neoplasm Recurrence, Local drug therapy, Oligopeptides therapeutic use, Oncolytic Virotherapy adverse effects, Oncolytic Viruses genetics
- Abstract
Purpose: Testing safety of Delta24-RGD (DNX-2401), an oncolytic adenovirus, locally delivered by convection enhanced delivery (CED) in tumor and surrounding brain of patients with recurrent glioblastoma., Patients and Methods: Dose-escalation phase I study with 3+3 cohorts, dosing 107 to 1 × 1011 viral particles (vp) in 20 patients. Besides clinical parameters, adverse events, and radiologic findings, blood, cerebrospinal fluid (CSF), brain interstitial fluid, and excreta were sampled over time and analyzed for presence of immune response, viral replication, distribution, and shedding., Results: Of 20 enrolled patients, 19 received the oncolytic adenovirus Delta24-RGD, which was found to be safe and feasible. Four patients demonstrated tumor response on MRI, one with complete regression and still alive after 8 years. Most serious adverse events were attributed to increased intracranial pressure caused by either an inflammatory reaction responding to steroid treatment or viral meningitis being transient and self-limiting. Often viral DNA concentrations in CSF increased over time, peaking after 2 to 4 weeks and remaining up to 3 months. Concomitantly Th1- and Th2-associated cytokine levels and numbers of CD3+ T and natural killer cells increased. Posttreatment tumor specimens revealed increased numbers of macrophages and CD4+ and CD8+ T cells. No evidence of viral shedding in excreta was observed., Conclusions: CED of Delta24-RGD not only in the tumor but also in surrounding brain is safe, induces a local inflammatory reaction, and shows promising clinical responses., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
6. Immunological responses to adjuvant vaccination with combined CD1c + myeloid and plasmacytoid dendritic cells in stage III melanoma patients.
- Author
-
Bloemendal M, Bol KF, Boudewijns S, Gorris MAJ, de Wilt JHW, Croockewit SAJ, van Rossum MM, de Goede AL, Petry K, Koornstra RHT, Figdor C, Gerritsen WR, Schreibelt G, and de Vries IJM
- Subjects
- Humans, CD8-Positive T-Lymphocytes, Dendritic Cells, Adjuvants, Immunologic, Vaccination, Glycoproteins, Antigens, CD1, Melanoma, Cutaneous Malignant, Cancer Vaccines therapeutic use, Melanoma therapy
- Abstract
We evaluated the immunological responses of lymph-node involved (stage III) melanoma patients to adjuvant dendritic cell vaccination with subsets of naturally occurring dendritic cells (nDCs). Fifteen patients with completely resected stage III melanoma were randomized to receive adjuvant dendritic cell vaccination with CD1c
+ myeloid dendritic cells (cDC2s), plasmacytoid dendritic cells (pDCs) or the combination. Immunological response was the primary endpoint and secondary endpoints included safety and survival. In 80% of the patients, antigen-specific CD8+ T cells were detected in skin test-derived T cells and in 55% of patients, antigen-specific CD8+ T cells were detectable in peripheral blood. Functional interferon-γ-producing T cells were found in the skin test of 64% of the patients. Production of nDC vaccines meeting release criteria was feasible for all patients. Vaccination only induced grade 1-2 adverse events, mainly consisting of fatigue. In conclusion, adjuvant dendritic cell vaccination with cDC2s and/or pDCs is feasible, safe and induced immunological responses in the majority of stage III melanoma patients., Competing Interests: No potential conflict of interest was reported by the author(s)., (© 2021 The Author(s). Published with license by Taylor & Francis Group, LLC.)- Published
- 2021
- Full Text
- View/download PDF
7. What does cell therapy manufacturing cost? A framework and methodology to facilitate academic and other small-scale cell therapy manufacturing costings.
- Author
-
Ten Ham RMT, Hövels AM, Hoekman J, Frederix GWJ, Leufkens HGM, Klungel OH, Jedema I, Veld SAJ, Nikolic T, Van Pel M, Zwaginga JJ, Lin F, de Goede AL, Schreibelt G, Budde S, de Vries IJM, Wilkie GM, Dolstra H, Ovelgönne H, Meij P, Mountford JC, Turner ML, and Hoefnagel MHN
- Subjects
- Commerce, Europe, Health Facilities, Humans, Academies and Institutes, Cell- and Tissue-Based Therapy economics, Costs and Cost Analysis
- Abstract
Background Aims: Recent technical and clinical advances with cell-based therapies (CBTs) hold great promise in the treatment of patients with rare diseases and those with high unmet medical need. Currently the majority of CBTs are developed and manufactured in specialized academic facilities. Due to small scale, unique characteristics and specific supply chain, CBT manufacturing is considered costly compared to more conventional medicinal products. As a result, biomedical researchers and clinicians are increasingly faced with cost considerations in CBT development. The objective of this research was to develop a costing framework and methodology for academic and other small-scale facilities that manufacture cell-based therapies., Methods: We conducted an international multi-center costing study in four facilities in Europe using eight CBTs as case studies. This study includes costs from cell or tissue procurement to release of final product for clinical use. First, via interviews with research scientists, clinicians, biomedical scientists, pharmacists and technicians, we designed a high-level costing framework. Next, we developed a more detailed uniform methodology to allocate cost items. Costs were divided into steps (tissue procurement, manufacturing and fill-finish). The steps were each subdivided into cost categories (materials, equipment, personnel and facility), and each category was broken down into facility running (fixed) costs and operational (variable) costs. The methodology was tested via the case studies and validated in developer interviews. Costs are expressed in 2018 euros (€)., Results: The framework and methodology were applicable across facilities and proved sensitive to differences in product and facility characteristics. Case study cost estimates ranged between €23 033 and €190 799 Euros per batch, with batch yield varying between 1 and 88 doses. The cost estimations revealed hidden costs to developers and provided insights into cost drivers to help design manufacturing best practices., Conclusions: This framework and methodology provide step-by-step guidance to estimate manufacturing costs specifically for cell-based therapies manufactured in academic and other small-scale enterprises. The framework and methodology can be used to inform and plan cost-conscious strategies for CBTs., (Copyright © 2020 International Society for Cell and Gene Therapy. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
8. Blood-derived dendritic cell vaccinations induce immune responses that correlate with clinical outcome in patients with chemo-naive castration-resistant prostate cancer.
- Author
-
Westdorp H, Creemers JHA, van Oort IM, Schreibelt G, Gorris MAJ, Mehra N, Simons M, de Goede AL, van Rossum MM, Croockewit AJ, Figdor CG, Witjes JA, Aarntzen EHJG, Mus RDM, Brüning M, Petry K, Gotthardt M, Barentsz JO, de Vries IJM, and Gerritsen WR
- Subjects
- Aged, Antigens, Neoplasm immunology, Humans, Kaplan-Meier Estimate, Male, Membrane Proteins immunology, Middle Aged, Mucin-1 immunology, Neoplasm Proteins immunology, Prostatic Neoplasms, Castration-Resistant immunology, Prostatic Neoplasms, Castration-Resistant mortality, Skin immunology, T-Lymphocytes immunology, Treatment Outcome, Vaccination adverse effects, Cancer Vaccines, Dendritic Cells immunology, Prostatic Neoplasms, Castration-Resistant therapy
- Abstract
Background: Clinical benefit of cellular immunotherapy has been shown in patients with castration-resistant prostate cancer (CRPC). We investigated the immunological response and clinical outcome of vaccination with blood-derived CD1c
+ myeloid dendritic cells (mDCs; cDC2) and plasmacytoid DCs (pDCs)., Methods: In this randomized phase IIa trial, 21 chemo-naive CRPC patients received maximally 9 vaccinations with mature mDCs, pDCs or a combination of mDCs plus pDCs. DCs were stimulated with protamine/mRNA and loaded with tumor-associated antigens NY-ESO-1, MAGE-C2 and MUC1. Primary endpoint was the immunological response after DC vaccination, which was monitored in peripheral blood and in T cell cultures of biopsies of post-treatment delayed-type hypersensitivity-skin tests. Main secondary endpoints were safety, feasibility, radiological PFS (rPFS) and overall survival. Radiological responses were assessed by MRIs and contrast-enhanced68 Ga-prostate-specific membrane antigen PET/CT, according to RECIST 1.1, PCWG2 criteria and immune-related response criteria., Results: Both tetramer/dextramer-positive (dm+ ) and IFN-γ-producing (IFN-γ+ ) antigen specific T cells were detected more frequently in skin biopsies of patients with radiological non-progressive disease (5/13 patients; 38%) compared to patients with progressive disease (0/8 patients; 0%). In these patients with vaccination enhanced dm+ and IFN-γ+ antigen-specific T cells median rPFS was 18.8 months (n = 5) vs. 5.1 months (n = 16) in patients without IFN-γ-producing antigen-specific T cells (p = 0.02). The overall median rPFS was 9.5 months. All DC vaccines were well tolerated with grade 1-2 toxicity., Conclusions: Immunotherapy with blood-derived DC subsets was feasible and safe and induced functional antigen-specific T cells. The presence of functional antigen-specific T cells correlated with an improved clinical outcome., Trial Registration: ClinicalTrials.gov identifier NCT02692976, registered 26 February 2016, retrospectively registered.- Published
- 2019
- Full Text
- View/download PDF
9. HIV-1 evolution in patients undergoing immunotherapy with Tat, Rev, and Nef expressing dendritic cells followed by treatment interruption.
- Author
-
de Goede AL, van Deutekom HW, Vrancken B, Schutten M, Allard SD, van Baalen CA, Osterhaus AD, Thielemans K, Aerts JL, Keşmir C, Lemey P, and Gruters RA
- Subjects
- Anti-Retroviral Agents therapeutic use, CD8-Positive T-Lymphocytes immunology, Dendritic Cells virology, Epitopes, T-Lymphocyte genetics, Evolution, Molecular, HIV Infections virology, HIV-1 classification, HIV-1 isolation & purification, Humans, RNA, Viral blood, RNA, Viral genetics, Sequence Analysis, DNA, nef Gene Products, Human Immunodeficiency Virus genetics, rev Gene Products, Human Immunodeficiency Virus genetics, tat Gene Products, Human Immunodeficiency Virus genetics, Dendritic Cells immunology, HIV Infections therapy, HIV-1 genetics, Immunotherapy methods, nef Gene Products, Human Immunodeficiency Virus immunology, rev Gene Products, Human Immunodeficiency Virus immunology, tat Gene Products, Human Immunodeficiency Virus immunology
- Abstract
Objectives: This study aimed to evaluate HIV sequence evolution in whole genes and in CD8 T-cell epitope regions following immunotherapy and subsequent analytical treatment interruption (ATI). A second objective of this study was to analyze associations between vaccine-specific immune responses and epitope mutation rates., Design: HIV-1-infected patients on combined antiretroviral therapy (cART) were subjected to immunotherapy by the administration of an autologous dendritic cell-based therapeutic vaccine expressing Tat, Rev, and Nef and subsequent ATI., Methods: HIV-1 genes were amplified and sequenced from plasma RNA obtained before initiation of cART as well as during ATI. Control sequences for virus evolution in untreated HIV-1-infected individuals were obtained from the HIV Sequence Database (Los Alamos). CD8 T-cell epitope regions were defined based on literature data and prediction models. HIV-1-specific immune responses were evaluated to analyze their impact on sequence evolution., Results: Viral sequence evolution in the tat, rev, and nef genes of vaccinated patients was similar to that of controls. The number of mutations observed inside and outside CD8 T-cell epitopes was comparable for vaccine-targeted and nontargeted proteins. We found no evidence for an impact of vaccine-induced or enhanced immune responses on the number of mutations inside or outside epitopes., Conclusion: Therapeutic vaccination of HIV-1-infected patients with a dendritic cell-based vaccine targeting Tat, Rev, and Nef did not affect virus evolution at the whole gene level nor at the CD8 T-cell epitope level.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.