Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maarten L. Zandvliet is active.

Publication


Featured researches published by Maarten L. Zandvliet.


Haematologica | 2010

Combined CD8+ and CD4+ adenovirus hexon-specific T cells associated with viral clearance after stem cell transplantation as treatment for adenovirus infection

Maarten L. Zandvliet; J.H. Frederik Falkenburg; Ellis van Liempt; Louise A. Veltrop-Duits; Arjan C. Lankester; Jayant S. Kalpoe; Michel G.D. Kester; Dirk M. van der Steen; Maarten J. D. van Tol; R. Willemze; Henk-Jan Guchelaar; Marco W. Schilham; Pauline Meij

Background Human adenovirus can cause morbidity and mortality in immunocompromised patients after allogeneic stem cell transplantation. Reconstitution of adenovirus-specific CD4+ T cells has been reported to be associated with sustained protection from adenovirus disease, but epitope specificity of these responses has not been characterized. Since mainly CD4+ T cells and no CD8+ T cells specific for adenovirus have been detected after allogeneic stem cell transplantation, the relative contribution of adenovirus-specific CD4+ and CD8+ T cells in protection from adenovirus disease remains to be elucidated. Design and Methods The presence of human adenovirus hexon-specific T cells was investigated in peripheral blood of pediatric and adult allogeneic stem cell transplant recipients, who showed spontaneous resolution of disseminated adenovirus infection. Subsequently, a clinical grade method was developed for rapid generation of adenovirus-specific T-cell lines for adoptive immunotherapy. Results Clearance of human adenovirus viremia coincided with emergence of a coordinated CD8+ and CD4+ T-cell response against adenovirus hexon epitopes in patients after allogeneic stem cell transplantation. Activation of adenovirus hexon-specific CD8+ and CD4+ T cells with a hexon protein-spanning peptide pool followed by interferon-γ-based isolation allowed rapid expansion of highly specific T-cell lines from healthy adults, including donors with very low frequencies of adenovirus hexon-specific T cells. Adenovirus-specific T-cell lines recognized multiple MHC class I and II restricted epitopes, including known and novel epitopes, and efficiently lysed human adenovirus-infected target cells. Conclusions This study provides a rationale and strategy for the adoptive transfer of donor-derived human adenovirus hexon-specific CD8+ and CD4+ T cells for the treatment of disseminated adenovirus infection after allogeneic stem cell transplantation.


Journal of Immunotherapy | 2011

Simultaneous isolation of CD8(+) and CD4(+) T cells specific for multiple viruses for broad antiviral immune reconstitution after allogeneic stem cell transplantation.

Maarten L. Zandvliet; Ellis van Liempt; Inge Jedema; Simone Kruithof; Michel G.D. Kester; Henk-Jan Guchelaar; J.H. Frederik Falkenburg; Pauline Meij

Opportunistic viral infections can cause serious morbidity and mortality in immunocompromised patients after allogeneic stem cell transplantation. Clinical studies have shown that adoptive transfer of donor-derived T cells specific for cytomegalovirus (CMV), Epstein-Barr virus (EBV), or human adenovirus (HAdV) can be a safe and effective treatment of infections with these major viral pathogens. The aim of this study was to develop a method for the simultaneous isolation of coordinated CD8+ and CD4+ memory T-cell responses against a broad repertoire of viral epitopes. To ensure that the method was applicable to a wide variety of virus-specific T cells that may differ in phenotypic and functional properties, we focused on T cells specific for the persistent viruses, CMV and EBV, and T cells specific for HAdV and influenza (FLU), which are not repetitively activated in vivo after initial viral clearance. Following in vitro activation, nearly all T cells specific for these viruses produced interferon &ggr; (IFN-&ggr;) and tumor necrosis factor &agr;, and expressed CD137, whereas the populations varied in the production of interleukin-2, degranulation, and expression of phenotypic markers. Different kinetics of IFN-&ggr; production were observed in CMV/EBV-specific T cells and HAdV/FLU-specific T cells. However, after the stimulation of peripheral blood from seropositive donors with viral protein-spanning peptide pools, the activated virus-specific CD8+ and CD4+ T cells could be simultaneously isolated by either IFN-&ggr;-based or CD137-based enrichment. This study provides an efficient and widely applicable strategy for the isolation of virus-specific T cells, which may be used for the reconstitution of virus-specific immunity in allogeneic stem cell transplantation recipients.


Cytotherapy | 2010

Co-ordinated isolation of CD8+ and CD4+ T cells recognizing a broad repertoire of cytomegalovirus pp65 and IE1 epitopes for highly specific adoptive immunotherapy

Maarten L. Zandvliet; Ellis van Liempt; Inge Jedema; Louise A. Veltrop-Duits; R. Willemze; Henk-Jan Guchelaar; J.H. Frederik Falkenburg; Pauline Meij

BACKGROUND AIMS Adoptive transfer of cytomegalovirus (CMV)-specific memory T cells can be used for treatment of CMV reactivation after allogeneic stem cell transplantation. As co-ordinated CD8(+) and CD4(+) T cells specific for a broad repertoire of CMV epitopes may be most effective for adoptive immunotherapy, the aim of this study was to isolate these cells from peripheral blood of CMV seropositive donors, irrespective of their HLA type. METHODS Activation of CMV-specific CD8(+) and CD4(+) T cells was compared after stimulation of donor peripheral blood with minimal epitope peptides, pools of overlapping 15-mer peptides or full-length protein. Furthermore, the kinetics of interferon (IFN)-γ production after stimulation was analyzed to determine the optimal time-point for IFN-γ-based isolation of CMV-specific T cells. The specificity, phenotype and functionality of generated T-cell lines were analyzed. RESULTS CMV protein-spanning 15-mer peptide pools induced simultaneous activation of both CD8(+) and CD4(+) CMV-specific T cells, while full-length CMV protein only efficiently activated CD4(+) CMV-specific T cells. Isolation of IFN-γ-secreting cells at the peak of the IFN-γ response after 4-h stimulation with CMV pp65 and IE1 peptide pools resulted in efficient enrichment of CMV-specific T cells. The T-cell lines contained high frequencies of CD8(+) and CD4(+) T cells recognizing multiple CMV pp65 and IE1 epitopes, and produced IFN-γ and tumor necrosis factor (TNF)-α upon specific restimulation. CONCLUSIONS This study provides a feasible strategy for the rapid generation of clinical-grade CD8(+) and CD4(+) T-cell lines with high specificity for multiple CMV pp65 and IE1 epitopes, which may be used for effective adoptive immunotherapy.


Journal of Immunotherapy | 2012

Effective Treatment of Refractory CMV Reactivation After Allogeneic Stem Cell Transplantation With In Vitro-generated CMV pp65-specific CD8+ T-cell Lines

Pauline Meij; Inge Jedema; Maarten L. Zandvliet; Pim L.J. van der Heiden; Marian van de Meent; H. M. Esther van Egmond; Ellis van Liempt; Conny Hoogstraten; Simone Kruithof; Sabrina A.J. Veld; Erik W.A. Marijt; Peter A. von dem Borne; Arjan C. Lankester; Constantijn J.M. Halkes; J.H. Frederik Falkenburg

To treat patients with refractory cytomegalovirus (CMV) reactivation after allogeneic stem cell transplantation, a phase I/II clinical study on adoptive transfer of in vitro-generated donor-derived or patient-derived CMV pp65-specific CD8+ T-cell lines was performed. Peripheral blood mononuclear cells from CMV seropositive donors or patients were stimulated with HLA-A*0201-restricted and/or HLA-B*0702-restricted CMV pp65 peptides (NLV/TPR) and 1 day after stimulation interferon-&ggr;)-producing cells were enriched using the CliniMACS Cytokine Capture System (interferon-&ggr;), and cultured with autologous feeders and low-dose interluekin-2. After 7–14 days of culture, quality controls were performed and the CMV-specific T-cell lines were administered or cryopreserved. The T-cell lines generated contained 0.6–17×106 cells, comprising 54%–96% CMV pp65-specific CD8+ T cells, and showed CMV-specific lysis of target cells. Fifteen CMV-specific T-cell lines were generated of which 8 were administered to patients with refractory CMV reactivation. After administration, no acute adverse events and no graft versus host disease were observed and CMV load disappeared. In several patients, a direct relation between administration of the T-cell line and the in vivo appearance of CMV pp65-specific T cells could be documented. In conclusion, administration of CMV pp65-specific CD8+ T-cell lines was found to be feasible and safe, and enduring efficacy of administered CMV pp65-specific CD8+ T-cell lines could be demonstrated.


Journal of Immunotherapy | 2009

Detailed analysis of IFNg response upon activation permits efficient isolation of cytomegalovirus-specific CD8+ T cells for adoptive immunotherapy.

Maarten L. Zandvliet; J.H. Frederik Falkenburg; Inge Jedema; R. Willemze; Henk-Jan Guchelaar; Pauline Meij

Adoptive transfer of donor-derived cytomegalovirus (CMV)-specific T cells may provide long-lived protection from CMV disease after allogeneic stem cell transplantation. Isolation of IFNg-secreting cells after CMV peptide stimulation can be performed by IFNg capture assay to generate highly specific T-cell lines without the need for extensive culture, which may hamper their in vivo efficacy. To exploit the full potential of this approach, we analyzed the IFNg response of CMV-specific CD8+ T cells in detail. Kinetic studies showed that T-cell receptor down-regulation coincided with the induction of IFNg production upon activation, which rapidly declined thereafter despite the continued presence of specific peptide. By varying the strength of stimulation we observed that overstimulation can result in profound T-cell receptor down-regulation, more rapid decline of IFNg production and reduced expansion. On the basis of these findings, we defined optimal conditions for IFNg-based isolation of CMV-specific CD8+ T cells with maximal potential for clinical application. These data stress the importance of analyses of the kinetics of cytokine production for isolation of T cells specific for other infectious or malignant antigens to exploit the full potential of cytokine capture isolation of antigen-specific T cells.


Oncotarget | 2016

TLR2 ligand-synthetic long peptide conjugates effectively stimulate tumor-draining lymph node T cells of cervical cancer patients.

Gijs G. Zom; Marij J. P. Welters; Nikki M. Loof; Renske Goedemans; Sinéad M. Lougheed; Rob Valentijn; Maarten L. Zandvliet; Nico J. Meeuwenoord; Cornelis J. M. Melief; Tanja D. de Gruijl; Gijsbert A. van der Marel; Dmitri V. Filippov; Ferry Ossendorp; Sjoerd H. van der Burg

The potency of human papillomavirus type 16 (HPV16)-encoded synthetic long peptides (SLP), conjugated to an optimized Toll-like receptor 2 ligand (TLR2-L), was assessed in ex vivo activation of HPV16+ cancer patient-derived T cells. Two highly immunogenic SLP sequences derived from the oncogenic E6 protein of HPV16 were selected and conjugated to a Pam3CSK4-based TLR2-L under GMP conditions. Both conjugates were able to mature human DCs in vitro and to activate human skin-derived antigen-presenting cells (APCs) upon intradermal injection in an ex vivo skin model, associated with induction of a favorable chemokine profile to attract and activate T cells. The conjugated SLPs were efficiently processed by APCs, since HPV16-specific CD4+ and CD8+ T-cell clones isolated from HPV16+ cervical tumors proliferated in response to both conjugates. The TLR2-L SLP conjugates significantly enhanced ex vivo T helper type 1 T-cell activation in cell suspensions obtained from tumor-draining lymph nodes (LN) resected during hysterectomy of HPV16+ cervical cancer patients. These results show that TLR2-L SLP conjugates can activate circulating or LN-derived tumor-specific T cells, a promising outcome for studying these two conjugates in a phase I/II clinical safety and immunogenicity trial.


Drug Discovery Today | 2016

Development of cell therapy medicinal products by academic institutes

Sofieke de Wilde; Henk-Jan Guchelaar; Carla Herberts; Mark W. Lowdell; Martin Hildebrandt; Maarten L. Zandvliet; Pauline Meij

In the rapidly evolving fields of cellular immunotherapy, gene therapy and regenerative medicine, a wide range of promising cell therapy medicinal products are in clinical development. Most products originate from academic research and are explored in early exploratory clinical trials. However, the success rate toward approval for regular patient care is disappointingly low. In this paper, we define strengths and hurdles applying to the development of cell therapy medicinal products in academic institutes, and analyze why only a few promising cell therapies have reached late-stage clinical development. Subsequently, we provide recommendations to stakeholders involved in development of cell therapies to exploit their potential clinical benefit.


British Journal of Clinical Pharmacology | 2017

A review of factors explaining variability in fentanyl pharmacokinetics; focus on implications for cancer patients

Evelien J. M. Kuip; Maarten L. Zandvliet; Stijn L.W. Koolen; Ron H.J. Mathijssen; Carin C.D. van der Rijt

Fentanyl is a strong opioid that is available for various administration routes, and which is widely used to treat cancer‐related pain. Many factors influence the fentanyl pharmacokinetics leading to a wide inter‐ and intrapatient variability. This systematic review summarizes multiple studied factors that potentially influence fentanyl pharmacokinetics with a focus on implications for cancer patients. The use of CYP3A4 inhibitors and inducers, impaired liver function, and heating of the patch potentially influence fentanyl pharmacokinetics in a clinically relevant way. In elderly patients, current data suggest that we should carefully dose fentanyl due to alterations in absorption and metabolism. The influence of BMI and gender on fentanyl pharmacokinetics is questionable, most probably due to a large heterogeneity in the published studies. Pharmacogenetics, e.g. the CYP3A5*3 gene polymorphism, may influence fentanyl pharmacokinetics as well, although further study is warranted. Several other factors have been studied but did not show significant and clinically relevant effects on fentanyl pharmacokinetics. Unfortunately, most of the published papers that studied factors influencing fentanyl pharmacokinetics describe healthy volunteers instead of cancer patients. Results from the studies in volunteers may not be simply extrapolated to cancer patients because of multiple confounding factors. To handle fentanyl treatment in a population of cancer patients, it is essential that physicians recognize factors that influence fentanyl pharmacokinetics, thereby preventing potential side‐effects and increasing its efficacy.


Cytotherapy | 2016

Hurdles in clinical implementation of academic advanced therapy medicinal products: A national evaluation

Sofieke de Wilde; Louise A. Veltrop-Duits; Merel Hoozemans-Strik; Thirza Ras; Janine Blom-Veenman; Henk-Jan Guchelaar; Maarten L. Zandvliet; Pauline Meij

BACKGROUND Since the implementation of the European Union (EU) regulation for advanced therapy medicinal products (ATMPs) in 2009, only six ATMPs achieved marketing authorization approval in the EU. Recognizing the major developments in the ATMP field, starting mostly in academic institutions, we investigated which hurdles were experienced in the whole pathway of ATMP development towards clinical care. METHODS Quality interviews were executed with different stakeholders in The Netherlands involved in the ATMP development field, e.g. academic research groups, national authorities and patient organizations. Based on the hurdles mentioned in the interviews, questionnaires were subsequently sent to the academic principal investigators (PIs) and ATMP good manufacturing practice (GMP) facility managers to quantify these hurdles. RESULTS Besides the familiar regulatory routes of marketing authorization (MA) and hospital exemption (HE), a part of the academic PIs perceived that ATMPs should become available by the Tissues and Cells Directive or did not anticipate on the next development steps towards implementation of their ATMP towards regular clinical care. The main hurdles identified were: inadequate financial support, rapidly evolving field, study-related problems, lacking regulatory knowledge, lack of collaborations and responsibility issues. DISCUSSION Creating an academic environment stimulating and planning ATMP development and licensing as well as investing in expanding the relevant regulatory knowledge in academic institutions seems a prerequisite to develop ATMPs from bench to patient.


Cytotherapy | 2017

Understanding clinical development of chimeric antigen receptor T cell therapies

Sofieke de Wilde; Henk-Jan Guchelaar; Maarten L. Zandvliet; Pauline Meij

BACKGROUND AIMS In the past decade, many clinical trials with gene- and cell-based therapies (GCTs) have been performed. Increased interest in the development of these drug products by various stakeholders has become apparent. Despite this growth in clinical studies, the number of therapies receiving marketing authorization approval (MAA) is lagging behind. To enhance the success rate of GCT development, it is essential to better understand the clinical development of these products. Chimeric antigen receptor (CAR) T cells are a GCT product subtype with promising efficacy in cancer treatment which are tested in many clinical trials, but have not yet received MAA. METHODS We generated an overview of the characteristics of CAR T-cell clinical development in the United States, Canada and Europe. Subsequently, the characteristics of clinical trials with CAR T-cell products that proceeded to a subsequent clinical trial, used as a proxy for success, were compared with those that did not proceed. RESULT From the U.S. and European Union clinical trial databases, 106 CAR T-cell trials were selected, from which 49 were linked to a subsequent trial and 57 were not. The majority of the trials had an academic sponsor from which most did not proceed, whereas most commercially sponsored trials were followed by another clinical trial. Furthermore, trials with a subsequent trial more frequently recruited large patient cohorts and were more often multicenter compared with trials that were not followed up. DISCUSSION These characteristics can be used by investigators to better design clinical trials with CAR T cells. We encourage sponsors to plan clinical development ahead for a higher efficiency of product development and thereby achieving a higher success rate of development towards MAA.

Collaboration


Dive into the Maarten L. Zandvliet's collaboration.

Top Co-Authors

Avatar

Pauline Meij

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Henk-Jan Guchelaar

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

J.H. Frederik Falkenburg

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Louise A. Veltrop-Duits

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ellis van Liempt

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Inge Jedema

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sofieke de Wilde

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

R. Willemze

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Arjan C. Lankester

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marco W. Schilham

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge