18 results on '"Smits, Evelien"'
Search Results
2. Trial watch: chemotherapy-induced immunogenic cell death in oncology.
- Author
-
Sprooten, Jenny, Laureano, Raquel S., Vanmeerbeek, Isaure, Govaerts, Jannes, Naulaerts, Stefan, Borras, Daniel M., Kinget, Lisa, Fucíková, Jitka, Špíšek, Radek, Jelínková, Lenka Palová, Kepp, Oliver, Kroemer, Guido, Krysko, Dmitri V., Coosemans, An, Vaes, Rianne D.W., De Ruysscher, Dirk, De Vleeschouwer, Steven, Wauters, Els, Smits, Evelien, and Tejpar, Sabine
- Subjects
CELL death ,TYPE I interferons ,CHEMOTHERAPY complications ,IMMUNE response ,CANCER cells - Abstract
Immunogenic cell death (ICD) refers to an immunologically distinct process of regulated cell death that activates, rather than suppresses, innate and adaptive immune responses. Such responses culminate into T cell-driven immunity against antigens derived from dying cancer cells. The potency of ICD is dependent on the immunogenicity of dying cells as defined by the antigenicity of these cells and their ability to expose immunostimulatory molecules like damage-associated molecular patterns (DAMPs) and cytokines like type I interferons (IFNs). Moreover, it is crucial that the host's immune system can adequately detect the antigenicity and adjuvanticity of these dying cells. Over the years, several well-known chemotherapies have been validated as potent ICD inducers, including (but not limited to) anthracyclines, paclitaxels, and oxaliplatin. Such ICD-inducing chemotherapeutic drugs can serve as important combinatorial partners for anti-cancer immunotherapies against highly immuno-resistant tumors. In this Trial Watch, we describe current trends in the preclinical and clinical integration of ICD-inducing chemotherapy in the existing immuno-oncological paradigms. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. Targeting CD70 in combination with chemotherapy to enhance the anti-tumor immune effects in non-small cell lung cancer.
- Author
-
Flieswasser, Tal, Van den Eynde, Astrid, Freire Boullosa, Laurie, Melis, Jöran, Hermans, Christophe, Merlin, Céline, Lau, Ho Wa, Van Audenaerde, Jonas, Lardon, Filip, Smits, Evelien, Pauwels, Patrick, and Jacobs, Julie
- Subjects
NON-small-cell lung carcinoma ,COMBINATION drug therapy ,IMMUNE checkpoint proteins ,IMMUNE checkpoint inhibitors ,KILLER cells - Abstract
Despite the recent emergence of immune checkpoint inhibitors, clinical outcomes of metastatic NSCLC patients remain poor, pointing out the unmet need to develop novel therapies to enhance the anti-tumor immune response in NSCLC. In this regard, aberrant expression of the immune checkpoint molecule CD70 has been reported on many cancer types, including NSCLC. In this study, the cytotoxic and immune stimulatory potential of an antibody-based anti-CD70 (aCD70) therapy was explored as single agent and in combination with docetaxel and cisplatin in NSCLC in vitro and in vivo. Anti-CD70 therapy resulted in NK-mediated killing of NSCLC cells and increased production of pro-inflammatory cytokines by NK cells in vitro. The combination of chemotherapy and anti-CD70 therapy further enhanced NSCLC cell killing. Moreover, in vivo findings showed that the sequential treatment of chemo-immunotherapy resulted in a significant improved survival and delayed tumor growth compared to single agents in Lewis Lung carcinoma-bearing mice. The immunogenic potential of the chemotherapeutic regimen was further highlighted by increased numbers of dendritic cells in the tumor-draining lymph nodes in these tumor-bearing mice after treatment. The sequential combination therapy resulted in enhanced intratumoral infiltration of both T and NK cells, as well as an increase in the ratio of CD8+ T cells over Tregs. The superior effect of the sequential combination therapy on survival was further confirmed in a NCI-H1975-bearing humanized IL15-NSG-CD34+ mouse model. These novel preclinical data demonstrate the potential of combining chemotherapy and aCD70 therapy to enhance anti-tumor immune responses in NSCLC patients. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. Trial watch: Dendritic cell (DC)-based immunotherapy for cancer.
- Author
-
Laureano, Raquel S., Sprooten, Jenny, Vanmeerbeerk, Isaure, Borras, Daniel M., Govaerts, Jannes, Naulaerts, Stefan, Berneman, Zwi N., Beuselinck, Benoit, Bol, Kalijn F., Borst, Jannie, Coosemans, an, Datsi, Angeliki, Fučíková, Jitka, Kinget, Lisa, Neyns, Bart, Schreibelt, Gerty, Smits, Evelien, Sorg, Rüdiger V., Spisek, Radek, and Thielemans, Kris
- Subjects
DENDRITIC cells ,IMMUNOTHERAPY ,CANCER vaccines ,IMMUNE response ,T cells - Abstract
Dendritic cell (DC)-based vaccination for cancer treatment has seen considerable development over recent decades. However, this field is currently in a state of flux toward niche-applications, owing to recent paradigm-shifts in immuno-oncology mobilized by T cell-targeting immunotherapies. DC vaccines are typically generated using autologous (patient-derived) DCs exposed to tumor-associated or -specific antigens (TAAs or TSAs), in the presence of immunostimulatory molecules to induce DC maturation, followed by reinfusion into patients. Accordingly, DC vaccines can induce TAA/TSA-specific CD8
+ /CD4+ T cell responses. Yet, DC vaccination still shows suboptimal anti-tumor efficacy in the clinic. Extensive efforts are ongoing to improve the immunogenicity and efficacy of DC vaccines, often by employing combinatorial chemo-immunotherapy regimens. In this Trial Watch, we summarize the recent preclinical and clinical developments in this field and discuss the ongoing trends and future perspectives of DCbased immunotherapy for oncological indications. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
5. Poly(I:C) primes primary human glioblastoma cells for an immune response invigorated by PD-L1 blockade.
- Author
-
Waele, Jorrit De, Marcq, Elly, Van Audenaerde, Jonas R. M., Loenhout, Jinthe Van, Deben, Christophe, Zwaenepoel, Karen, Kelft, Erik Van de, Planken, David Van der, Menovsky, Tomas, Van den Bergh, Johan M. J., Willemen, Yannick, Pauwels, Patrick, Berneman, Zwi N., Lardon, Filip, Peeters, Marc, Wouters, An, and Smits, Evelien L. J.
- Subjects
GLIOBLASTOMA multiforme ,IMMUNE response ,CANCER immunotherapy ,PROGNOSIS - Abstract
Prognosis of glioblastoma remains dismal, underscoring the need for novel therapies. Immunotherapy is generating promising results, but requires combination strategies to unlock its full potential. We investigated the immunomodulatory capacities of poly(I:C) on primary human glioblastoma cells and its combinatorial potential with programmed death ligand (PD-L) blockade. In our experiments, poly(I:C) stimulated expression of both PD-L1 and PD-L2 on glioblastoma cells, and a pro-inflammatory secretome, including type I interferons (IFN) and chemokines CXCL9, CXCL10, CCL4 and CCL5. IFN-α was partially responsible for the elevated PD-1 ligand expression on these cells. Moreover, real-time PCR and chloroquine-mediated blocking experiments indicated that poly(I:C) triggered Toll-like receptor 3 to elicit its effect. Cocultures of poly(I:C)- treated glioblastoma cells with peripheral blood mononuclear cells enhanced lymphocytic activation (CD69, IFN-γ) and cytotoxic capacity (CD107a, granzyme B). Additional PD-L1 blockade further propagated immune activation. Besides activating immunity, poly(I:C)-treated glioblastoma cells also doubled the attraction of CD8
+ T cells, and to a lesser extent CD4+ T cells, via a mechanism which included CXCR3 and CCR5 ligands. Our results indicate that by triggering glioblastoma cells, poly(I:C) primes the tumor microenvironment for an immune response. Secreted cytokines allow for immune activation while chemokines attract CD8C T cells to the front, which are postulated as a prerequisite for effective PD-1/PD-L1 blockade. Accordingly, additional blockade of the concurrently elevated tumoral PD-L1 further reinforces the immune activation. In conclusion, our data proposes poly(I:C) treatment combined with PD-L1 blockade to invigorate the immune checkpoint inhibition response in glioblastoma. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
6. Generation of a cord blood-derived Wilms Tumor 1 dendritic cell vaccine for AML patients treated with allogeneic cord blood transplantation.
- Author
-
de Haar, Colin, Plantinga, Maud, Blokland, Nina J. G., van Til, Niek P., Flinsenberg, Thijs W. H., Van Tendeloo, Viggo F., Smits, Evelien L., Boon, Louis, Spel, Lotte, Boes, Marianne, Boelens, Jaap Jan, and Nierkens, Stefan
- Subjects
NEPHROBLASTOMA ,DENDRITIC cells ,CORD blood transplantation ,ACUTE myeloid leukemia treatment ,IMMUNOTHERAPY - Abstract
The poor survival rates of refractory/relapsed acute myeloid leukemia (AML) patients after haematopoietic cell transplantation (HCT) requires the development of additional immune therapeutic strategies. As the elicitation of tumor-antigen specific cytotoxic T lymphocytes (CTLs) is associated with reduced relapses and enhanced survival, enhanced priming of these CTLs using an anti-AML vaccine may result in long-term immunity against AML. Cord blood (CB), as allogeneic HCT source, may provide a unique setting for such post-HCT vaccination, considering its enhanced graft-versus-leukemia (GvL) effects and population of highly responsive naïve T cells. It is our goal to develop a powerful and safe immune therapeutic strategy composed of CB-HCT followed by vaccination with CB CD34
C -derived dendritic cells (DCs) presenting the oncoprotein Wilms Tumor-1 (WT1), which is expressed in AML-blasts in the majority of patients. Here, we describe the optimization of a clinically applicable DC culture protocol. This two-step protocol consisting of an expansion phase followed by the differentiation toward DCs, enables us to generate sufficient cord blood-derived DCs (CBDCs) in the clinical setting. At the end of the culture, the CBDCs exhibit a mature surface phenotype, are able to migrate, express tumor antigen (WT1) after electroporation with mRNA encoding the full-length WT1 protein, and stimulate WT1-specific T cells. [ABSTRACT FROM AUTHOR]- Published
- 2015
- Full Text
- View/download PDF
7. Plasma, blood and liver tissue sample preparation methods for the separate quantification of liposomal-encapsulated prednisolone phosphate and non-encapsulated prednisolone.
- Author
-
Smits, Evelien A. W., Soetekouw, José A., Bakker, Peter F. A., Baijens, Bart J. H., and Vromans, Herman
- Subjects
- *
PREDNISOLONE , *ENCAPSULATION (Catalysis) , *LIVER analysis , *TISSUE engineering , *TISSUE analysis , *BLOOD plasma , *BLOOD sampling - Abstract
Besides the development of sample preparation methods for the determination of separate liposomal-encapsulated prednisolone phosphate and non-encapsulated prednisolone concentrations in murine plasma and blood, this article also presents the first description of an accurate sample preparation method for the determination of such separate concentrations in the murine liver. The quantitative differentiation is based on the immediate hydrolysis of prednisolone phosphate (PP) into prednisolone (P) after its release from the liposomes in vivo: PP represents the encapsulated drug, while P represents the non-encapsulated drug. The use of 10 ml methanol/g tissue during homogenization of liver tissue ensures complete liposome rupture, prevention of the dephosphorylation of PP released during homogenization, sufficient clean supernatants, excellent extraction of P and sufficient extraction of PP and excellent accuracies and precision complying with the internal guidelines for pre-clinical studies (80-120% and maximal 20%, respectively). Similarly, the matching sample preparation methods for plasma and blood involve protein precipitation with four equivalents of methanol also ensuring accuracies and precision complying with the internal guidelines for pre-clinical studies. Application of these sample preparation methods is going to generate the first pharmacokinetic (PK) profile of a liposomal preparation, in which the encapsulated and non-encapsulated drug concentrations in a tissue are measured separately. Such separated concentration profiles can gain important insights into the PKs of liposomal PP and probably also with regard to liposomal formulations in general, like the quantification of the in vivo drug release from the liposomes. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
8. Immunotherapy: is a minor god yet in the pantheon of treatments for lung cancer?
- Author
-
Rolfo, Christian, Sortino, Giovanni, Smits, Evelien, Passiglia, Francesco, Bronte, Giuseppe, Castiglia, Marta, Russo, Antonio, Santos, Edgardo S, Janssens, Annelies, Pauwels, Patrick, and Raez, Luis
- Abstract
Immunotherapy has been studied for many years in lung cancer without significant results, making the majority of oncologists quite skeptical about its possible application for non-small cell lung cancer treatment. However, the recent knowledge about immune escape and subsequent 'cancer immunoediting' has yielded the development of new strategies of cancer immunotherapy, heralding a new era of lung cancer treatment. Cancer vaccines, including both whole-cell and peptide vaccines have been tested both in early and advanced stages of non-small cell lung cancer. New immunomodulatory agents, including anti-CTLA4, anti-PD1/PDL1 monoclonal antibodies, have been investigated as monotherapy in metastatic lung cancer. To date, these treatments have shown impressive results of efficacy and tolerability in early clinical trials, leading to testing in several large, randomized Phase III trials. As these results will be confirmed, these drugs will be available in the near future, offering new exciting therapeutic options for lung cancer treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
9. In vitro confirmation of the quantitative differentiation of liposomal encapsulated and non-encapsulated prednisolone (phosphate) tissue concentrations by murine phosphatases.
- Author
-
Smits, Evelien A. W., Soetekouw, José A., and Vromans, Herman
- Subjects
- *
PREDNISOLONE , *LIPOSOMES , *DEPHOSPHORYLATION , *PRODRUGS , *PHOSPHATASES , *EXCRETORY organs , *PHARMACOKINETICS - Abstract
The quantitative differentiation of liposomal encapsulated and non-encapsulated drug tissue concentrations is desirable, since the efficacy and toxicity are only related to the level of non- encapsulated drug. However, such separate concentration profiles in tissues have still not been reported due to lacking analytical methodology. The encapsulation of prodrugs like prednisolone phosphate (PP) in liposomes offers new, analytical opportunities. Instantaneous dephosphorylation of PP into prednisolone (P) by phosphatases after its release from the liposome in vivo makes it possible to differentiate between the encapsulated and the non- encapsulated drug for such preparations of liposomal PP: PP represents the encapsulated drug, while P represents the non-encapsulated drug. In the here described study, the instantaneous dephosphorylation of PP by murine liver and kidney phosphatases has been verified by incubation of PP in liver and kidney homogenates followed by estimation of the dephosphorylation rate constants k and the dephosphorylation time of the expected maximal in vivo non-encapsulated drug concentrations. In vitro PP has been rapidly converted into P in the presence of homogenate from the excretory organs. The calculated values for k have shown that the liver contains more active sites per gram of tissue than the kidneys. However, the dephosphorylation of PP by these active sites is slower compared with the kidneys. Compared with other pharmacokinetic processes of P, the estimated dephosphorylation times of the expected maximal in vivo non-encapsulated drug concentrations in the liver and the kidneys are considered to be instantaneous. This enables the separate determination of the encapsulated and non-encapsulated drug concentrations in the excretory organs after administration of liposomal PP in mice generating the first pharmacokinetic profile of a liposomal preparation, in which the in vivo encapsulated and free drug tissues concentrations are measured separately. This can also gain important insights into the pharmacokinetics of liposomal formulations in general. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
10. Interleukin-15 dendritic cells as vaccine candidates for cancer immunotherapy.
- Author
-
Anguille, Sébastien, Lion, Eva, Vanden Bergh, Johan, Van Acker, Heleen H., Willemen, Yannick, Smits, Evelien L., Van Tendeloo, Viggo F., and Berneman, Zwi N.
- Published
- 2013
- Full Text
- View/download PDF
11. Dendritic cell vaccination in acute myeloid leukemia.
- Author
-
Anguille, Sébastien, Willemen, Yannick, Lion, Eva, Smits, Evelien L., and Berneman, Zwi N.
- Subjects
ACUTE myeloid leukemia ,CANCER patients ,DRUG therapy ,IMMUNOTHERAPY ,DENDRITIC cells ,TRANSPLANTATION of organs, tissues, etc. - Abstract
The prognosis of patients with acute myeloid leukemia (AML) remains dismal, with a 5-year overall survival rate of only 5.2% for the continuously growing subgroup of AML patients older than 65 years. These patients are generally not considered eligible for intensive chemotherapy and/or allogeneic hematopoietic stem cell transplantation because of high treatment-related morbidity and mortality, emphasizing the need for novel, less toxic, treatment alternatives. It is within this context that immunotherapy has gained attention in recent years. In this review, we focus on the use of dendritic cell (DC) vaccines for immunotherapy of AML. DC are central orchestrators of the immune system, bridging innate and adaptive immunity and critical to the induction of anti-leukemic immunity. We discuss the rationale and basic principles of DC-based therapy for AML and review the clinical experience that has been obtained so far with this form of immunotherapy for patients with AML. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
12. Monofluorinated unsymmetrical bent‐core mesogens.
- Author
-
Achten, Remko, Smits, Evelien A. W., Reddy, R. Amaranatha, Giesbers, Marcel, Marcelis, Antonius T. M., and Sudhölter, Ernst J. R.
- Subjects
- *
FLUORINE , *BENDING (Metalwork) , *ORGANIC compounds , *RESORCINOL , *ESTERS , *MICROSCOPY , *CALORIMETRY , *X-ray diffraction - Abstract
The synthesis and mesomorphic properties of 30 bent-core compounds with a fluorine substituent in one of the outer rings are reported. The banana-shaped compounds are all derived from resorcinol and contain esters as linking groups between the five aromatic rings. The different mesophases have been characterized by polarizing optical microscopy, differential scanning calorimetry, X-ray diffraction studies and electro-optical investigations. The compounds with the longer terminal chains exhibit an antiferroelectric SmCP phase. Upon introduction of a fluorine substituent the layer spacing increases, as compared with the corresponding unsubstituted compound. The introduction of one terminal vinyl group in the mono-substituted bent-core mesogens has no significant influence on the liquid crystalline properties and the switching remains antiferroelectric. Due to the introduction of the terminal double bond, these banana-shaped compounds are suitable for the preparation of siloxane polymers or attachment to a hydrogen-terminated silicon surface. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
13. Interferon α may be back on track to treat acute myeloid leukemia.
- Author
-
Smits, Evelien L. J. M., Anguille, Sébastien, and Berneman, Zwi N.
- Subjects
- *
INTERFERONS , *ANTIVIRAL agents , *ANTINEOPLASTIC agents , *ACUTE myeloid leukemia treatment , *ACUTE leukemia - Abstract
Our own experience and a thorough literature review suggest that interferon α (IFNα) should be reconsidered for the treatment of acute myeloid leukemia patients. Most likely, the success of such treatment depends on the achievement of high serum levels of IFNα for several months, which can be obtained by using pegylated IFNα. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
14. CD56 marks human dendritic cell subsets with cytotoxic potential.
- Author
-
Roothans, Dessie, Smits, Evelien, Lion, Eva, Tel, Jurjen, and Anguille, Sébastien
- Subjects
- *
DENDRITIC cells , *KILLER cells , *CELL-mediated cytotoxicity , *TUMOR antigens , *TUMOR markers , *CD5 antigen , *CYTOTOXIC T cells - Abstract
Human plasmacytoid and myeloid dendritic cells (DCs), when appropriately stimulated, can express the archetypal natural killer (NK)-cell surface marker CD56. In addition to classical DC functions, CD56+ DCs are endowed with an unconventional cytotoxic capacity. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
15. Unveiling a CD70-positive subset of cancer-associated fibroblasts marked by pro-migratory activity and thriving regulatory T cell accumulation.
- Author
-
Jacobs, Julie, Deschoolmeester, Vanessa, Zwaenepoel, Karen, Flieswasser, Tal, Deben, Christophe, Van den Bossche, Jolien, Hermans, Christophe, Rolfo, Christian, Peeters, Marc, De Wever, Olivier, Lardon, Filip, Siozopoulou, Vasiliki, Smits, Evelien, and Pauwels, Patrick
- Subjects
COLON cancer ,FIBROBLASTS ,T cells - Abstract
Cancer-associated fibroblasts (CAFs) are involved in the proliferative and invasive behavior of colorectal cancer (CRC). Nonetheless, CAFs represent a heterogeneous population with both cancer-promoting and cancer-restraining actions, lacking specific markers to target them. Expression of the immune checkpoint molecule CD70 is normally limited to cells of the lymphoid lineage. Instead, tumor cells hijack CD70 to facilitate immune evasion by increasing the amount of suppressive regulatory T cells (Tregs). The aim of this study was to explore CD70 expression patterns in CRC, not merely focusing on the tumor cells, but also taking the tumor stromal cells into account. We have analyzed the prognostic value of CD70 expression by immunohistochemistry in CRC specimens and its relationship with well-known fibroblast markers and Tregs. In addition, in vitro experiments were conducted to unravel the role of CD70-positive CAFs on migration and immune escape. We reveal prominent expression of CD70 on a specific subset of CAFs in invasive CRC specimens. Cancer cells show almost no expression of CD70. The presence of CD70-positive CAFs proved to be an independent adverse prognostic marker. Functionally, CD70-positive CAFs stimulated migration and significantly increased the frequency of naturally occurring Tregs. In conclusion, we have identified the expression of CD70 on CAFs as a novel prognostic marker for CRC. We have found evidence of a cross talk between CD70
+ CAFs and naturally occurring Tregs, paving the way towards immune escape. As such, this study provides a strong rationale for the exploration of CD70-targeting antibodies in CRC. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
16. Prognostic and predictive aspects of the tumor immune microenvironment and immune checkpoints in malignant pleural mesothelioma.
- Author
-
Marcq, Elly, Siozopoulou, Vasiliki, De Waele, Jorrit, van Audenaerde, Jonas, Zwaenepoel, Karen, Santermans, Eva, Hens, Niel, Pauwels, Patrick, van Meerbeeck, Jan P., and Smits, Evelien L. J.
- Subjects
CANCER prognosis ,IMMUNE system ,MESOTHELIOMA ,THERAPEUTICS - Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer with a poor prognosis and an increasing incidence, for which novel therapeutic strategies are urgently required. Since the immune system has been described to play a presumed role in the protection against MPM, characterization of its tumor immune microenvironment (TME) and immune checkpoints can identify new immunotherapeutic targets and their predictive and/or prognostic value. To characterize the TME and the immune checkpoint expression profile, we performed immunohistochemistry (IHC) on formalin-fixed paraffin embedded (FFPE) tissue sections from 54 MPM patients (40 at time of diagnosis; 14 treated with chemotherapy). We stained for PD-1, PD-L1, TIM-3, LAG-3, CD4, CD8, CD45RO, granzyme B, FoxP3 and CD68. Furthermore, we analyzed the relationship between the immunological parameters and survival, as well as response to chemotherapy. We found that TIM-3, PD-1 and PD-L1 were expressed on both immune and tumor cells. Strikingly, PD-1 and PD-L1 expression on tumor cells was only seen in unpretreated samples. No LAG-3 expression was observed. CD45RO expression in the stroma was an independent negative predictive factor for response on chemotherapy, while CD4 and TIM-3 expression in lymphoid aggregates were independent prognostic factors for better outcome. Our data propose TIM-3 as a promising new target in mesothelioma. Chemotherapy influences the expression of immune checkpoints and therefore further research on the best combination treatment schedule is required. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
17. Viral infections following allogeneic stem cell transplantation: how to cure the cure?
- Author
-
Smits, Evelien L. J. and Berneman, Zwi N.
- Subjects
- *
VIRUS diseases , *STEM cell transplantation , *CYTOMEGALOVIRUS diseases , *POLYOMAVIRUSES , *DRUG therapy , *RADIATION , *IMMUNOTHERAPY - Abstract
The article reports on a study related to primary viral infections and latent viruses including cytomegalovirus (CMV) and polyomavirus BK (BKV), published within this issue. It states that the conditioning regimen, consisting of chemotherapy and radiation, removes virus-specific memory T cells and creates a permissive environment for latent viruses to replicate. However, it provides insight into the potential for immunotherapy to stimulate virus-specific immune recovery.
- Published
- 2010
- Full Text
- View/download PDF
18. Human blood myeloid and plasmacytoid dendritic cells cross activate each other and synergize in inducing NK cell cytotoxicity.
- Author
-
van Beek, Jasper J. P., Gorris, Mark A. J., Sköld, Annette E., Hatipoglu, Ibrahim, Van Acker, Heleen H., Smits, Evelien L., de Vries, I. Jolanda M., and Bakdash, Ghaith
- Subjects
CELLULAR immunity ,CANCER immunotherapy ,CANCER treatment ,BIOLOGICAL crosstalk ,KILLER cells - Abstract
Human blood dendritic cells (DCs) hold great potential for use in anticancer immunotherapies. CD1c+myeloid DCs and plasmacytoid DCs (pDCs) have been successfully utilized in clinical vaccination trials against melanoma. We hypothesize that combining both DC subsets in a single vaccine can further improve vaccine efficacy. Here, we have determined the potential synergy between the two subsetsin vitroon the level of maturation, cytokine expression, and effector cell induction. Toll-like receptor (TLR) stimulation of CD1c+DCs induced cross-activation of immature pDCs and vice versa. When both subsets were stimulated together using TLR agonists, CD86 expression on pDCs was increased and higher levels of interferon (IFN)-α were produced by DC co-cultures. Although the two subsets did not display any synergistic effect on naive CD4+and CD8+T cell polarization, CD1c+DCs and pDCs were able to complement each other's induction of other immune effector cells. The mere presence of pDCs in DC co-cultures promoted plasma cell differentiation from activated autologous B cells. Similarly, CD1c+DCs, alone or in co-cultures, induced high levels of IFN-γ from allogeneic peripheral blood lymphocytes or activated autologous natural killer (NK) cells. Both CD1c+DCs and pDCs could enhance NK cell cytotoxicity, and interestingly DC co-cultures further enhanced NK cell-mediated killing of an NK-resistant tumor cell line. These results indicate that co-application of human blood DC subsets could render DC-based anticancer vaccines more efficacious. [ABSTRACT FROM PUBLISHER]
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.