16 results on '"Robbert Van Der Voort"'
Search Results
2. Improving dendritic cell vaccine immunogenicity by silencing PD-1 ligands using siRNA-lipid nanoparticles combined with antigen mRNA electroporation
- Author
-
Jamie Wong, Stuart Milstein, Willemijn Hobo, Robbert van der Voort, Tatiana Novobrantseva, Hila Epstein-Barash, Nicolaas Schaap, Harry Dolstra, Ju Liu, and Hanny Fredrix
- Subjects
Cancer Research ,T cell ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Immunology ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,Transfection ,Cancer Vaccines ,Antigen ,Antigens, Neoplasm ,Translational research [ONCOL 3] ,medicine ,Humans ,Immunology and Allergy ,Gene Silencing ,RNA, Messenger ,RNA, Small Interfering ,Immune Regulation Translational research [NCMLS 2] ,Gene knockdown ,Immunogenicity ,Electroporation ,Dendritic Cells ,Immunotherapy ,Dendritic cell ,Programmed Cell Death 1 Ligand 2 Protein ,Lipids ,Molecular biology ,medicine.anatomical_structure ,Oncology ,Leukocytes, Mononuclear ,Cancer research ,Nanoparticles - Abstract
Item does not contain fulltext Dendritic cell (DC)-based vaccination boosting antigen-specific immunity is being explored for the treatment of cancer and chronic viral infections. Although DC-based immunotherapy can induce immunological responses, its clinical benefit has been limited, indicating that further improvement of DC vaccine potency is essential. In this study, we explored the generation of a clinical-grade applicable DC vaccine with improved immunogenic potential by combining PD-1 ligand siRNA and target antigen mRNA delivery. We demonstrated that PD-L1 and PD-L2 siRNA delivery using DLin-KC2-DMA-containing lipid nanoparticles (LNP) mediated efficient and specific knockdown of PD-L expression on human monocyte-derived DC. The established siRNA-LNP transfection method did not affect DC phenotype or migratory capacity and resulted in acceptable DC viability. Furthermore, we showed that siRNA-LNP transfection can be successfully combined with both target antigen peptide loading and mRNA electroporation. Finally, we demonstrated that these PD-L-silenced DC loaded with antigen mRNA superiorly boost ex vivo antigen-specific CD8(+) T cell responses from transplanted cancer patients. Together, these findings indicate that our PD-L siRNA-LNP-modified DC are attractive cells for clinical-grade production and in vivo application to induce and boost immune responses not only in transplanted cancer patients, but likely also in other settings. 01 februari 2013
- Published
- 2012
- Full Text
- View/download PDF
3. PD-1/PD-L1 interactions contribute to functional T-cell impairment in patients who relapse with cancer after allogeneic stem cell transplantation
- Author
-
Alan J. Korman, Robbert van der Voort, Theo de Witte, J.H. Frederik Falkenburg, Michel G.D. Kester, Frans Maas, Harry Dolstra, Nicolaas Schaap, Wieger J. Norde, Michael Quigley, Willemijn Hobo, and Konnie M. Hebeda
- Subjects
Cancer Research ,T cell ,Programmed Cell Death 1 Receptor ,T-Cell Antigen Receptor Specificity ,Biology ,B7-H1 Antigen ,Minor Histocompatibility Antigens ,Interleukin 21 ,Interferon-gamma ,Cancer stem cell ,Translational research [ONCOL 3] ,Antigens, CD ,Recurrence ,T-Lymphocyte Subsets ,chronic viral-infection programmed death-1 myeloid-leukemia tumor exhaustion expression pathway pd-1 immunotherapy vaccination ,medicine ,Cytotoxic T cell ,Humans ,Transplantation, Homologous ,IL-2 receptor ,Interleukin 3 ,CD86 ,Inflammation ,Immune Regulation Translational research [NCMLS 2] ,Gene Expression Regulation, Leukemic ,Tumor Necrosis Factor-alpha ,Gene Expression Profiling ,Translational research Immune Regulation [ONCOL 3] ,Hematopoietic Stem Cell Transplantation ,Coculture Techniques ,Neoplasm Proteins ,medicine.anatomical_structure ,Oncology ,Leukemia, Myeloid ,Immunology ,B7-1 Antigen ,Neoplastic Stem Cells ,Tumor Escape ,B7-2 Antigen ,Apoptosis Regulatory Proteins ,Receptors, Purinergic P2X5 ,Immunologic Memory ,CD80 - Abstract
Contains fulltext : 97194.pdf (Publisher’s version ) (Closed access) Tumor relapses remain a serious problem after allogeneic stem cell transplantation (alloSCT), despite the long-term persistence of minor histocompatibility antigen (MiHA)-specific memory CD8(+) T cells specific for the tumor. We hypothesized that these memory T cells may lose their function over time in transplanted patients. Here, we offer functional and mechanistic support for this hypothesis, based on immune inhibition by programmed death-1 (PD-1) expressed on MiHA-specific CD8(+) T cells and the associated role of the PD-1 ligand PD-L1 on myeloid leukemia cells, especially under inflammatory conditions. PD-L1 was highly upregulated on immature human leukemic progenitor cells, whereas costimulatory molecules such as CD80 and CD86 were not expressed. Thus, immature leukemic progenitor cells seemed to evade the immune system by inhibiting T-cell function via the PD-1/PD-L1 pathway. Blocking PD-1 signaling using human antibodies led to elevated proliferation and IFN-gamma production of MiHA-specific T cells cocultured with PD-L1-expressing leukemia cells. Moreover, patients with relapsed leukemia after initial MiHA-specific T-cell responses displayed high PD-L1 expression on CD34(+) leukemia cells and increased PD-1 levels on MiHA-specific CD8(+) T cells. Importantly, blocking PD-1/PD-L1 interactions augment proliferation of MiHA-specific CD8(+) memory T cells from relapsed patients. Taken together, our findings indicate that the PD-1/PD-L pathway can be hijacked as an immune escape mechanism in hematological malignancies. Furthermore, they suggest that blocking the PD-1 immune checkpoint offers an appealing immunotherapeutic strategy following alloSCT in patients with recurrent or relapsed disease.
- Published
- 2011
- Full Text
- View/download PDF
4. Intratumoral rhIL-12 administration in head and neck squamous cell carcinoma patients induces B cell activation
- Author
-
Robbert van der Voort, Gosse J. Adema, I. Jolanda M. de Vries, Ina S. Klasen, Aniek O. de Graaf, Tjitske Duiveman-de Boer, Jeroen van der Laak, Ruurd Torensma, Johan H. J. M. van Krieken, Léon C van Kempen, Harry Dolstra, Carla M.L. van Herpen, and Pieter H.M. De Mulder
- Subjects
Male ,Cancer Research ,Pathology ,Genetics and epigenetic pathways of disease [NCMLS 6] ,medicine.medical_treatment ,Injections, Intralesional ,Lymphocyte Activation ,Polymerase Chain Reaction ,Immunophenotyping ,Immune Regulation [NCMLS 2] ,Molecular diagnosis, prognosis and monitoring [UMCN 1.2] ,Chronic inflammation and autoimmunity [UMCN 4.2] ,B-Lymphocytes ,Middle Aged ,Immunohistochemistry ,Interleukin-12 ,Recombinant Proteins ,Pathogenesis and modulation of inflammation [N4i 1] ,Cytokine ,medicine.anatomical_structure ,Oncology ,Head and Neck Neoplasms ,Carcinoma, Squamous Cell ,Female ,Lymph ,medicine.medical_specialty ,Age-related aspects of cancer [ONCOL 2] ,Interferon-gamma ,Translational research [ONCOL 3] ,Carcinoma ,medicine ,Humans ,RNA, Messenger ,B cell ,Aged ,DNA Primers ,Hereditary cancer and cancer-related syndromes [ONCOL 1] ,Base Sequence ,business.industry ,Tumor-infiltrating lymphocytes ,Germinal center ,Immunotherapy, gene therapy and transplantation [UMCN 1.4] ,Tissue engineering and pathology [NCMLS 3] ,medicine.disease ,Head and neck squamous-cell carcinoma ,Tumor microenvironment [UMCN 1.3] ,Lymph Nodes ,business ,Immunity, infection and tissue repair [NCMLS 1] - Abstract
Contains fulltext : 69631.pdf (Publisher’s version ) (Open Access) The objectives of this study were to investigate the effects of intratumorally (i.t.) administered recombinant human interleukin-12 (rhIL-12) on the distribution and function of B cells in the primary tumors, the locoregional lymph nodes and peripheral blood of head and neck squamous cell carcinoma (HNSCC) patients. The initial characterization of the patients participating in the phase Ib and phase II studies has previously been reported. After rhIL-12 treatment, fewer secondary follicles with a broader outer region of the mantle zones and an increase in interfollicular B-blasts were seen in the enlarged lymph nodes compared with control HNSCC patients. The size of the germinal center (GC) was diminished, partly due to a decrease in the number of CD57+ GC cells that have been associated with immune suppression. These changes did not correlate with signs of apoptosis or CXCR5 expression by B cells. Strikingly, in 3 out of 4 IL-12 treated patients, increased IFN-gamma mRNA expression by B cells was detected. In addition, a highly significant IgG subclass switch was seen in the plasma with more IgG1, less IgG2 and more IgG4, indicating a switch to T helper 1 phenotype. Finally, peritumoral B cell infiltration was a positive prognostic sign for overall survival in the 30 HNSCC patients investigated, irrespective of IL-12 treatment. In conclusion, these data indicate that after i.t. IL-12 treatment in HNSCC, significant activation of the B cell and the B cell compartment occurred and that the presence of tumor infiltrating B cells correlated with overall survival of HNSCC patients.
- Published
- 2008
5. Efficient nontoxic delivery of PD-L1 and PD-L2 siRNA into dendritic cell vaccines using the cationic lipid SAINT-18
- Author
-
Robbert van der Voort, Willemijn Hobo, Harry Dolstra, Kasper Teijgeler, Wieger J. Norde, Nicolaas Schaap, Hanny Fredrix, Mieke W H Roeven, and Marcel H. J. Ruiters
- Subjects
Cancer Research ,Small interfering RNA ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Immunology ,Pyridinium Compounds ,CD8-Positive T-Lymphocytes ,Transfection ,Cancer Vaccines ,B7-H1 Antigen ,Minor Histocompatibility Antigens ,Neoplasms ,Minor histocompatibility antigen ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,Transplantation, Homologous ,RNA, Small Interfering ,Cell Proliferation ,Pharmacology ,Gene knockdown ,Chemistry ,Electroporation ,Graft vs Tumor Effect ,Hematopoietic Stem Cell Transplantation ,Dendritic cell ,Dendritic Cells ,Programmed Cell Death 1 Ligand 2 Protein ,Molecular biology ,Clone Cells ,Transplantation ,Cancer research ,Cytokines ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] - Abstract
Dendritic cell (DC)-based vaccination is an appealing strategy to boost graft-versus-tumor immunity after allogeneic stem cell transplantation (allo-SCT), and thereby prevent or counteract tumor recurrence. By exploiting minor histocompatibility antigens (MiHA) presented on hematopoietic cells, donor CD8 T-cell immunity can be selectively targeted to patient's hematological tumor cells without the risk of inducing graft-versus-host disease. Previously, we demonstrated that silencing RNA (siRNA) of programmed death-ligand 1 (PD-L1) and PD-L2 on DCs markedly augments the expansion and function of MiHA-specific CD8 T cells. However, previously applied methods based on electroporation or lipid nanoparticles were either incompatible with target antigen mRNA delivery or required complex manufacturing compliant to Good Manufacturing Practice. Here, we investigated whether transfection using lipoplexes composed of PD-L1 and PD-L2 siRNAs plus SAINT-18:DOPE (ie, SAINT-RED) is an effective and feasible clinical-grade method in DC vaccine manufacturing. We observed that a single siRNA/SAINT-RED transfection resulted in efficient and long-term knockdown of the PD-1 ligands without affecting DC maturation or viability. Furthermore, we demonstrated that SAINT-RED can be heat sterilized without loss of function, facilitating its use in aseptic DC vaccine production. Finally, we showed that the established transfection method can be combined with target antigen mRNA or peptide loading to efficiently stimulate MiHA-specific T-cell expansion and cytokine production. Together, these findings indicate that the developed PD-L siRNA/SAINT-RED transfection protocol in combination with MiHA mRNA or peptide loading can be applied in the generation of clinical-grade DC vaccines to boost antitumor immunity after allo-SCT.
- Published
- 2015
6. siRNA silencing of PD-1 ligands on dendritic cell vaccines boosts the expansion of minor histocompatibility antigen-specific CD8(+) T cells in NOD/SCID/IL2Rg(null) mice
- Author
-
Nicolaas Schaap, Robbert van der Voort, Hanny Fredrix, Harry Dolstra, Willemijn Hobo, and Anniek B. van der Waart
- Subjects
Adoptive cell transfer ,Cancer Research ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,T cell ,Immunology ,Priming (immunology) ,Mice, SCID ,CD8-Positive T-Lymphocytes ,PD-L ,Biology ,Lymphocyte Activation ,Cancer Vaccines ,B7-H1 Antigen ,Minor Histocompatibility Antigens ,Mice ,Mice, Inbred NOD ,medicine ,Animals ,Cytotoxic T cell ,Immunology and Allergy ,Minor histocompatibility antigen ,RNA, Small Interfering ,Antigen-presenting cell ,Mice, Knockout ,Leukemia ,Dendritic Cells ,Dendritic cell ,Programmed Cell Death 1 Ligand 2 Protein ,Adoptive Transfer ,Coculture Techniques ,Adoptive cell therapy ,medicine.anatomical_structure ,Oncology ,Hematologic Neoplasms ,GVT ,RNA Interference ,Original Article ,CD8 ,Ex vivo ,Interleukin Receptor Common gamma Subunit - Abstract
Allogeneic stem cell transplantation (allo-SCT) can be a curative therapy for patients suffering from hematological malignancies. The therapeutic efficacy is based on donor-derived CD8+ T cells that recognize minor histocompatibility antigens (MiHAs) expressed by patient’s tumor cells. However, these responses are not always sufficient, and persistence and recurrence of the malignant disease are often observed. Therefore, application of additive therapy targeting hematopoietic-restricted MiHAs is essential. Adoptive transfer of MiHA-specific CD8+ T cells in combination with dendritic cell (DC) vaccination could be a promising strategy. Though effects of DC vaccination in anti-cancer therapy have been demonstrated, improvement in DC vaccination therapy is needed, as clinical responses are limited. In this study, we investigated the potency of program death ligand (PD-L) 1 and 2 silenced DC vaccines for ex vivo priming and in vivo boosting of MiHA-specific CD8+ T cell responses. Co-culturing CD8+ T cells with MiHA-loaded DCs resulted in priming and expansion of functional MiHA-specific CD8+ T cells from the naive repertoire, which was augmented upon silencing of PD-L1 and PD-L2. Furthermore, DC vaccination supported and expanded adoptively transferred antigen-specific CD8+ T cells in vivo. Importantly, the use of PD-L silenced DCs improved boosting and further expansion of ex vivo primed MiHA-specific CD8+ T cells in immunodeficient mice. In conclusion, adoptive transfer of ex vivo primed MiHA-specific CD8+ T cells in combination with PD-L silenced DC vaccination, targeting MiHAs restricted to the hematopoietic system, is an interesting approach to boost GVT immunity in allo-SCT patients and thereby prevent relapse. Electronic supplementary material The online version of this article (doi:10.1007/s00262-015-1668-6) contains supplementary material, which is available to authorized users.
- Published
- 2015
7. Efficient activation of LRH-1-specific CD8+T-cell responses from transplanted leukaemia patients by stimulation with P2X5 mRNA-electroporated dendritic cells
- Author
-
Theo de Witte, J.H. Frederik Falkenburg, Robbert van der Voort, Ingrid M. Overes, Michel G.D. Kester, Harry Dolstra, and Hanny Fredrix
- Subjects
Cancer Research ,Graft-vs-Leukemia Effect ,medicine.medical_treatment ,Immunology ,Graft vs Leukemia Effect ,Biology ,CD8-Positive T-Lymphocytes ,Cell Degranulation ,Minor Histocompatibility Antigens ,Interferon-gamma ,Immune system ,Immune Regulation [NCMLS 2] ,Translational research [ONCOL 3] ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,RNA, Messenger ,Antigen-presenting cell ,Pharmacology ,Receptors, Purinergic P2 ,Dendritic cell ,Immunotherapy ,Dendritic Cells ,Leukemia, Myeloid, Acute ,Electroporation ,Cancer research ,Stem cell ,Receptors, Purinergic P2X5 ,CD8 ,Stem Cell Transplantation - Abstract
Contains fulltext : 80821.pdf (Publisher’s version ) (Closed access) Alloreactive CD8+ T cells targeting minor histocompatibility antigens (MiHA) on malignant cells of the recipient play a pivotal role in graft-versus-tumor responses observed after allogeneic stem cell transplantation and donor lymphocyte infusion (DLI). However, these MiHA-specific CD8+ T-cell responses do not result in complete eradication of tumor cells in all patients. Furthermore, CD8+ memory T cells persisting after DLI do not always efficiently expand with recurrence of the disease. Adjuvant immunotherapy using dendritic cells (DC) loaded with hematopoietic-restricted MiHA may boost antitumor CD8+ T-cell immunity without inducing graft-versus-host disease. Here, we explored the use of mRNA-electroporated DC to stimulate MiHA-specific CD8+ T-cell responses. We demonstrate that electroporation of mature DC with P2X5 mRNA encoding for hematopoietic-restricted MiHA LRH-1 results in high expression of both mRNA and protein, and has no negative effect on the mature phenotype and migratory capacity of the DC. Furthermore, these DC can efficiently stimulate LRH-1-specific CD8+ effector T cells to proliferate and produce interferon-gamma. In addition, LRH-1-specific CD8+ memory T cells that are present in patient-derived peripheral blood mononuclear cells at long periods post-DLI can be effectively activated by stimulation with P2X5 mRNA-electroporated DC to proliferate and degranulate upon target cell recognition. These results indicate that adjuvant immunotherapy using DC electroporated with mRNA encoding hematopoietic-restricted MiHA mismatched between patients and donors may enhance the graft versus tumor response induced by stem cell transplantation and DLI.
- Published
- 2010
8. siRNA silencing of PD-L1 and PD-L2 on dendritic cells augments expansion and function of minor histocompatibility antigen-specific CD8+ T cells
- Author
-
Nicolaas Schaap, Robbert van der Voort, Harry Dolstra, Frans Maas, Willemijn Hobo, Theo de Witte, and Niken Adisty
- Subjects
Immunology ,CD8-Positive T-Lymphocytes ,Biology ,Biochemistry ,B7-H1 Antigen ,Minor Histocompatibility Antigens ,Antigen ,Antigens, CD ,Immune Regulation [NCMLS 2] ,Translational research [ONCOL 3] ,Minor histocompatibility antigen ,Humans ,Cytotoxic T cell ,Gene Silencing ,Antigen-presenting cell ,Cells, Cultured ,Cell Proliferation ,Dendritic Cells ,Cell Biology ,Hematology ,Dendritic cell ,Programmed Cell Death 1 Ligand 2 Protein ,Mixed lymphocyte reaction ,Cancer research ,Cytokines ,Intercellular Signaling Peptides and Proteins ,CD8 - Abstract
Contains fulltext : 87316.pdf (Publisher’s version ) (Closed access) Tumor relapse after human leukocyte antigen-matched allogeneic stem cell transplantation (SCT) remains a serious problem, despite the long-term presence of minor histocompatibility antigen (MiHA)-specific memory T cells. Dendritic cell (DC)-based vaccination boosting MiHA-specific T-cell immunity is an appealing strategy to prevent or counteract tumor recurrence, but improvement is necessary to increase the clinical benefit. Here, we investigated whether knockdown of programmed death ligand 1 (PD-L1) and PD-L2 on monocyte-derived DCs results in improved T-cell activation. Electroporation of single siRNA sequences into immature DCs resulted in efficient, specific, and long-lasting knockdown of PD-L1 and PD-L2 expression. PD-L knockdown DCs strongly augmented interferon-gamma and interleukin-2 production by stimulated T cells in an allogeneic mixed lymphocyte reaction, whereas no effect was observed on T-cell proliferation. Moreover, we demonstrated that PD-L gene silencing, especially combined PD-L1 and PD-L2 knockdown, resulted in improved proliferation and cytokine production of keyhole limpet hemocyanin-specific CD4(+) T cells. Most importantly, PD-L knockdown DCs showed superior potential to expand MiHA-specific CD8(+) effector and memory T cells from leukemia patients early after donor lymphocyte infusion and later during relapse. These data demonstrate that PD-L siRNA electroporated DCs are highly effective in enhancing T-cell proliferation and cytokine production, and are therefore attractive cells for improving the efficacy of DC vaccines in cancer patients.
- Published
- 2010
9. Aberrant expression of the hematopoietic-restricted minor histocompatibility antigen LRH-1 on solid tumors results in efficient cytotoxic T cell-mediated lysis
- Author
-
T Henriëtte Levenga, Harry Dolstra, Ingrid M. Overes, Robbert van der Voort, Johanna C. M. Vos, Pieter H.M. De Mulder, Theo de Witte, and Agnes van Horssen-Zoetbrood
- Subjects
Cancer Research ,Genotype ,Hematopoietic System ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Immunology ,CD8-Positive T-Lymphocytes ,Biology ,Minor Histocompatibility Antigens ,Immune Regulation [NCMLS 2] ,Translational research [ONCOL 3] ,Neoplasms ,medicine ,Minor histocompatibility antigen ,Humans ,Transplantation, Homologous ,Immunology and Allergy ,Cytotoxic T cell ,RNA, Messenger ,Lymphokine-activated killer cell ,Cardiovascular diseases [NCEBP 14] ,Receptors, Purinergic P2 ,Cancer ,Immunotherapy ,Intercellular Adhesion Molecule-1 ,medicine.disease ,DNA-Binding Proteins ,medicine.anatomical_structure ,Microscopy, Fluorescence ,Oncology ,Cancer research ,Stem cell ,Receptors, Purinergic P2X5 ,CD8 ,Stem Cell Transplantation ,Transcription Factors - Abstract
Contains fulltext : 79614.pdf (Publisher’s version ) (Closed access) CD8(+) T cells recognizing minor histocompatibility antigens (MiHA) on solid tumor cells may mediate effective graft-versus-tumor (GVT) reactivity after allogeneic stem cell transplantation (SCT). Previously, we identified LRH-1 as a hematopoietic-restricted MiHA encoded by the P2X5 gene. Here, we report that LRH-1 is aberrantly expressed on solid tumor cells. P2X5 mRNA expression is demonstrated in a significant portion of solid tumor cell lines, including renal cell carcinoma (RCC), melanoma, colorectal carcinoma, brain cancer and breast cancer. Importantly, P2X5 gene expression was also detected in a subset of primary solid tumor specimens derived from RCC, brain cancer and breast cancer patients. Furthermore, P2X5 expressing solid tumor cells can be effectively targeted by LRH-1-specific cytotoxic T lymphocytes under inflammatory conditions. The expression of HLA-B7 and CD54 on tumor cells increases upon cytokine stimulation resulting in improved T cell activation as observed by higher levels of degranulation and enhanced tumor cell lysis. Overall, hematopoietic-restricted MiHA LRH-1 is aberrantly expressed on solid tumor cells and may be used as target in GVT-specific immunotherapy after SCT.
- Published
- 2009
10. Combined IL-15 and IL-12 drives the generation of CD34+-derived natural killer cells with superior maturation and alloreactivity potential following adoptive transfer
- Author
-
Robbert van der Voort, Marleen Tordoir, Jan Spanholtz, Harry Dolstra, Nicolaas Schaap, Anniek B. van der Waart, Jeannette Cany, and Joop H. Jansen
- Subjects
Adoptive cell transfer ,Lymphokine-activated killer cell ,Immunology ,CD34 ,Biology ,Natural killer T cell ,Natural killer cell ,Interleukin 21 ,medicine.anatomical_structure ,Oncology ,Interleukin 15 ,medicine ,Cancer research ,Interleukin 12 ,Immunology and Allergy - Abstract
Adoptive transfer of allogeneic natural killer (NK) cells represents a promising treatment approach against cancer, including acute myeloid leukemia (AML). Previously, we reported a cytokine-based culture method for the generation of NK cell products with high cell number and purity. In this system, CD34+ hematopoietic progenitor cells (HPC) were expanded and differentiated into NK cells under stroma-free conditions in the presence of IL-15 and IL-2. We show that combining IL-15 with IL-12 drives the generation of more mature and highly functional NK cells. In particular, replacement of IL-2 by IL-12 enhanced the cytolytic activity and IFNγ production of HPC-NK cells toward cultured and primary AML cells in vitro, and improved antileukemic responses in NOD/SCID-IL2Rγnull (NSG) mice bearing human AML cells. Phenotypically, IL-12 increased the frequency of HPC-NK cells expressing NKG2A and killer immunoglobulin-like receptor (KIR), which were more responsive to target cell stimulation. In addition, NK15/12 ...
- Published
- 2015
- Full Text
- View/download PDF
11. In situ tumor ablation creates an antigen source for the generation of antitumor immunity
- Author
-
Roger P.M. Sutmuller, Gosse J. Adema, Carl G. Figdor, Erik Bennink, Theo J.M. Ruers, Robbert van der Voort, and Martijn H. den Brok
- Subjects
Male ,In situ ,Cancer Research ,Adoptive cell transfer ,medicine.drug_class ,T-Lymphocytes ,Melanoma, Experimental ,Biology ,Lymphocyte Activation ,Monoclonal antibody ,Immunotherapy, Adoptive ,Interferon-gamma ,Mice ,Immune system ,Antigen ,Antigens, CD ,Antigens, Neoplasm ,Immunity ,Cricetinae ,medicine ,Splenocyte ,Animals ,Cytotoxic T cell ,CTLA-4 Antigen ,Antibodies, Monoclonal ,Immunotherapy, gene therapy and transplantation [UMCN 1.4] ,Antigens, Differentiation ,Mice, Inbred C57BL ,Oncology ,Immunology ,Catheter Ablation ,Female - Abstract
Tumor-destructing techniques, like radiofrequency ablation (RFA), allow eradication of large tumors. Potentially, in situ tumor destruction also can provide the immune system with an antigen source for the induction of antitumor immunity. Antigen-presenting cells could take up antigens in the periphery after which they induce specific immune responses. Recent data show that especially antigen-presenting dendritic cells are crucial for the induction of potent immune responses. However, virtually nothing is known regarding the induction of immune responses after in situ tumor destruction in mice or humans. We used the well-defined murine B16-OVA melanoma cell line to develop a novel tumor model to explore: (a) the immunologic consequences of in situ tumor destruction; and (b) the efficacy of a combination approach of tumor destruction and immunostimulation. Applying this model system we demonstrate that following RFA, a weak but detectable immune response develops, directed against OVA, but also against a broader range of B16 antigens. Adoptive transfer experiments further indicate that antitumor reactivity can be transferred to naïve mice by splenocytes. To augment the response observed, we administered a blocking monoclonal antibody against cytotoxic T-lymphocyte-associated antigen 4 at the time of tumor destruction. Interestingly, this strongly enhanced antitumor immunity, resulting in long-lasting tumor protection. These results illustrate that in situ tumor destruction can provide a useful antigen source for the induction of antitumor immunity, provided that additional immunostimulatory signals are coadministered.
- Published
- 2004
12. Expression of c-Met and heparan-sulfate proteoglycan forms of CD44 in colorectal cancer
- Author
-
Esther A. Beuling, V. J. M. Wielenga, S. T. Pals, Marcel Spaargaren, Cees van Krimpen, Robbert van der Voort, L. Smit, T. E. I. Taher, and Other departments
- Subjects
C-Met ,(Patho)Physiological, endocrinological and methabolic aspects [Prevention of disorders in human reproduction] ,Colorectal cancer ,Mouse model of colorectal and intestinal cancer ,medicine.disease_cause ,Ligands ,Proto-Oncogene Mas ,Receptor tyrosine kinase ,Pathology and Forensic Medicine ,chemistry.chemical_compound ,medicine ,Humans ,(Patho-)fysiologische, endocriene en metabole aspecten. [Preventie van stoornissen in de menselijke voortplanting] ,Phosphorylation ,biology ,Hepatocyte Growth Factor ,CD44 ,Heparan sulfate ,Proto-Oncogene Proteins c-met ,medicine.disease ,Prognosis ,Hyaluronan Receptors ,chemistry ,biology.protein ,Cancer research ,Hepatocyte growth factor ,Carcinogenesis ,Colorectal Neoplasms ,Tumorimmunology ,Heparan Sulfate Proteoglycans ,medicine.drug ,Regular Articles - Abstract
In colorectal cancer patients, prognosis is not determined by the primary tumor but by the formation of distant metastases. Molecules that have been implicated in the metastatic process are the proto-oncogene product c-Met and CD44 glycoproteins. Recently, we obtained evidence for functional collaboration between these two molecules: CD44 isoforms decorated with heparan sulfate chains (CD44-HS) can bind the c-Met ligand, the growth and motility factor hepatocyte growth factor/scatter factor (HGF/SF). This interaction strongly promotes signaling through the receptor tyrosine kinase c-Met. In the present study, we explored the expression of CD44-HS, c-Met, and HGF/SF in the normal human colon mucosa, and in colorectal adenomas and carcinomas, as well as their interaction in colorectal cancer cell lines. Compared to the normal colon, CD44v3 isoforms, which contain a site for HS attachment, and c-Met, were both overexpressed on the neoplastic epithelium of colorectal adenomas and on most carcinomas. Likewise, HGF/SF was expressed at increased levels in tumor tissue. On all tested colorectal cancer cell lines CD44v3 and c-Met were co-expressed. As was shown by immunoprecipitation and Western blotting, CD44 on these cells lines was decorated with HS. Interaction with HS moieties on colorectal carcinoma (HT29) cells promoted HGF/SF-induced activation of c-Met and of the Ras-MAP kinase pathway. Interestingly, survival analysis showed that CD44-HS expression predicts unfavorable prognosis in patients with invasive colorectal carcinomas. Taken together, our findings indicate that CD44-HS, c-Met, and HGF/SF are simultaneously overexpressed in colorectal cancer and that HS moieties promote c-Met signaling in colon carcinoma cells. These observations suggest that collaboration between CD44-HS and the c-Met signaling pathway may play an important role in colorectal tumorigenesis.
- Published
- 2000
13. Akt Signalling Inhibition Promotes The Ex Vivo generation Of Minor Histocompatibility Antigen-Specific CD8+ Memory Stem T Cells
- Author
-
Harry Dolstra, Willemijn Hobo, Noortje van der Weem, Robbert van der Voort, Luca Gattinoni, Nicolaas Schaap, and Anniek B. van der Waart
- Subjects
ZAP70 ,T cell ,Immunology ,Cell Biology ,Hematology ,Biology ,Natural killer T cell ,Biochemistry ,Interleukin 21 ,medicine.anatomical_structure ,Cancer research ,medicine ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Interleukin 3 - Abstract
Allogeneic hematopoietic stem cell transplantation (allo-SCT) followed by donor lymphocyte infusion (DLI) is a potential curative treatment for patients suffering from a hematological malignancy. Efficacy is attributed to the graft-versus-tumor (GVT) response, during which engrafted donor T cells become activated by recipient minor histocompatibility antigens (MiHA) presented on dendritic cells (DC). Subsequently, these activated T cells expand, acquire effector functions and kill MiHA-positive tumor cells. However, persistence and recurrence of malignant disease is often observed, indicating that insufficient GVT immunity is induced. This imperfect alloreactive response is probably due to insufficient numbers of MiHA-specific effector T cells and/or defective antigen-presentation and costimulation. Therefore, adoptive transfer of potent ex vivo-generated MiHA-specific T cells, restricted to the hematopoietic system, would boost the GVT-effect without increasing the risk for GVHD. Although successful in vitro induction of MiHA-specific CD8+ T cells from naive precursors has been reported, the resulting antigen-experienced T cell population consist of fully differentiated effector-memory T cells (TEM). Over the past years it has been described that this T cell subset is not the most potent memory subset in anti-tumor responses in vivo following T cell transfer. In this regard, the less-differentiated memory subset called stem cell memory T cells (TSCM) with superior in vivo expansion, self-renewal capacity and plasticity to differentiate in potent effectors would generate a stronger GVT response. In this study, we aimed to investigate the in vivo availability and ex vivo generation of TSCM-like MiHA-specific T cells as additive treatment option for allo-SCT patients. First, we investigated whether in allo-SCT patients MiHA-specific T cells could be detected with a TSCM phenotype defined by the expression of CD45RO, CCR7, CD27 and CD95. Though TSCM cells could be clearly detected within CMV-specific CD8+ T cells in allo-SCT patients, similar to healthy controls, no MiHA-specific TSCM cells could be detected. This emphasises the need for more potent adoptive MiHA-specific T cell therapy following allo-SCT. Therefore, we next explored the possibility of generating TSCM-like CD8+ T cells by interfering with the Akt signalling pathway. Emerging findings indicate that the differentiation program of CD8+ T cells is dictated by the strength and duration of AKT activity. Therefore, we explored whether the pharmacological inhibition of this signaling pathway could results in the generation of TSCM-like CD8+ T cells. We stimulated CCR7+CD45RA+ naive CD8+ T cells with CD3/CD28 beads plus IL-2, IL-7 and/or IL-15 in the presence an Akt inhibitor. Interestingly, CD8+ T cells in these Akt-cultures were inhibited in their differentiation stage, expressing higher levels of CD45RA and CCR7 compared to controls. In addition, expression of CD95, IL2Rβ, and IL7Rα was also elevated confirming the TSCM-like phenotype. Although proliferation of the Akt-inhibited CD8+ T cells was decreased as shown by less PBSE dilution, expansion could be significantly preserved. Next, we investigated whether the established culture conditions could be used to generate MiHA-specific TSCM-like cells. Therefore, CD8+ T cells from MiHA-negative donors were primed using autologous MiHA peptide-loaded moDCs in the presence of the Akt-inhibitor. Interestingly, MiHA-specific T cell priming could be induced, consisting of mainly TCM and TSCM-like cells compared to almost entirely TEM cells in the control setting. Akt-inhibited MiHA-specific T cells showed higher expression of CCR7, CD45RA, CD62L, CD28, CD95, and IL7Rα. Importantly, for the Akt-inhibited MiHA-specific T cells, proliferation was reserved, resulting in robust proliferation capacity during restimulation after removal of the Akt-inhibitor. The resulting TEFF cells were highly functional, showing capacity to degranulate and produce IFNγ upon peptide restimulation. In conclusion, by inhibiting the Akt-pathway, in vitro CD8+ T cell differentiation can be reduced. Therefore, Akt signalling inhibition can be exploited for generating TSCM-like MiHA-specific T cells in adoptive immunotherapy after allo-SCT. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2013
- Full Text
- View/download PDF
14. Aberrant Expression in Human Epithelial Cancers of the P2X5-Encoded Minor Histocompatibility Antigen LRH-1: Implications for Graft-Versus-Tumor Immunity Against Solid Tumors
- Author
-
Harry Dolstra, Johanna C. M. Vos, T. de Witte, Henriette Levenga, Ingrid M. Overes, Pieter H.M. De Mulder, Agnes van Horssen-Zoetbrood, and Robbert van der Voort
- Subjects
Lymphokine-activated killer cell ,medicine.diagnostic_test ,medicine.medical_treatment ,Melanoma ,Immunology ,Cell Biology ,Hematology ,Immunotherapy ,Biology ,medicine.disease ,Biochemistry ,Flow cytometry ,CTL ,Cell culture ,medicine ,Cancer research ,Minor histocompatibility antigen ,Stem cell - Abstract
Allogeneic stem cell transplantation (SCT) in combination with donor lymphocyte infusion (DLI) is an experimental treatment for patients with metastatic solid tumors. The therapeutic efficacy is attributed to the graft-versus-tumor (GVT) response during which donor-derived T cells eliminate malignant cells via recognition of minor histocompatibility antigens (MiHA). To reduce accompanying GVHD, it is crucial to identify MiHA which are selectively expressed on hematopoietic cells and solid tumor cells. Previously, we identified a hematopoietic cell-restricted MiHA, designated LRH-1, which is presented by HLA-B7 and encoded by the P2X5 purinergic receptor gene (J. Clin. Invest.2005:115:3506–3516). Here, we report that LRH-1, in addition to its hematopoietic cell-restricted expression, is aberrantly expressed on epithelial tumor cell lines. We observed that P2X5 mRNA is significantly expressed in 14 out of 42 (33%) solid tumor cell lines tested by real-time quantitative RT-PCR. We detected P2X5 transcripts in 3 out of 11 renal cell carcinoma cell lines, 2 out of 4 melanoma cell lines, 3 out of 7 colorectal carcinoma cell lines, 4 out of 10 brain tumor cell lines and 2 out of 10 breast cancer cell lines. To determine whether P2X5 mRNA expression in solid tumor cell lines results in susceptibility to lysis by LRH-1-specific CTL, we performed flow cytometry-based cytotoxicity assays using P2X5-expressing tumor cell lines. Based on LRH-1 genotyping analysis, we selected six solid tumor cell lines for the cytotoxicity studies. Remarkably, LRH-1-specific CTL efficiently lysed and inhibited the growth of DAOY brain tumor cells up to 3 days of co-culture. The renal cell carcinoma cell lines SKRC-33 and SKRC-18 and the melanoma cell line BLM were also susceptible to LRH-1 CTL-mediated lysis, although less effectively. However, pre-incubation of these tumor cell lines with IFNγ and TNFα significantly increased the susceptibility to LRH-1-specific CTL and resulted in complete target cell lysis. Furthermore, these cytokine-stimulated cell lines induced higher levels of CTL degranulation as determined by CD107a staining. No cytotoxicity was observed against LRH-1-negative FM3 melanoma and SKRC-24 renal cell carcinoma cell lines. These findings illustrate that the GVT reactivity observed in solid tumors after allogeneic SCT may be selectively enhanced by LRH-1-specific immunotherapy.
- Published
- 2007
- Full Text
- View/download PDF
15. Human CD34+ Myeloid Leukemic Progenitor Cells Are Susceptible to Lysis by Minor Histocompatibility Antigen LRH-1-Specific Cytotoxic T Lymphocytes
- Author
-
Annemieke Vos, Anton Schattenberg, Ingrid M. Overes, Robbert van der Voort, Harry Dolstra, Inge Jedema, T. de Witte, Wieger J. Norde, J.H. Frederik Falkenburg, and Agnes van Horssen-Zoetbrood
- Subjects
Myeloid ,Immunology ,CD34 ,Cell Biology ,Hematology ,Biology ,Biochemistry ,CTL ,Haematopoiesis ,medicine.anatomical_structure ,Antigen ,hemic and lymphatic diseases ,Cancer research ,medicine ,Cytotoxic T cell ,Progenitor cell ,Stem cell - Abstract
Allogeneic stem cell transplantation (SCT) is a specialized form of immunotherapy for treating patients with hematological malignancies. The curative potential is attributed to the graft-versus-tumor (GVT) response during which donor-derived cytotoxic T lymphocytes (CTL) eliminate malignant cells of the recipient. Minor histocompatibility antigens (MiHA) are the major targets of the GVT response, and expansion of MiHA-specific CTL has been shown to coincide with tumor remission following SCT. Recently, we identified a novel hematopoietic cell-restricted MiHA, designated LRH-1, which is presented by HLA-B7 and encoded by the P2X5 purinergic receptor gene (J. Clin. Invest.2005:115:3506–3516). Interestingly, tetramer analysis showed a direct association between in vivo expansion of LRH-1-specific CD8+ T cells and the disappearance of Bcr-Abl positive tumor cells in the CML patient from whom LRH-1-specific CTL was originally isolated. In addition, we detected in vivo expansion of LRH-1-specific CTL in an AML patient who was in clinical remission without GVHD. Furthermore, we demonstrated that P2X5 mRNA is significantly expressed in leukemic CD34+ progenitor cells from most CML as well as AML patients. These findings indicate a role for LRH-1 in inducing GVT immunity against myeloid leukemic progenitor cells. Here, we investigated the ex vivo responsiveness of myeloid leukemic CD34+ progenitor cells to LRH-1-specific CTL. First, we addressed this question using the CD34+ KG1 cell line which is positive for LRH-1. By using a CFSE-based survival assay we demonstrated that KG1 cells stably transfected with HLA-B7 could be efficiently lysed by LRH-1-specific CTL. Next, we determined responsiveness of purified CD34+ progenitor cells from HLA-B7+ CML patients to LRH-1 CTL-mediated killing. In the CFSE-based survival assay as well as a hematopoietic progenitor cell inhibition assay we showed that LRH-1-specific CTL efficiently recognize and kill CD34+ progenitor cells from LRH-1+ CML patients. In contrast, LRH-1-specific CTL did not inhibit the proliferation of CD34+ progenitor cells from LRH-1- CML patients. These findings illustrate that the P2X5-encoded LRH-1 antigen is an attractive target for adequate eradication of myeloid leukemic progenitor cells after allogeneic SCT.
- Published
- 2006
- Full Text
- View/download PDF
16. Ex Vivo Generation Of Functional Plasmacytoid and Myeloid Dendritic Cells Is Strongly Promoted By The Aryl Hydrocarbon Receptor Antagonist Stemregenin 1
- Author
-
Marta Cossu, Robbert van der Voort, Jan Spanholtz, Soley Thordardottir, Tim J. A. Hutten, Hangalapura Basav N., Timothy R D J Radstake, Nicolaas Schaap, and Harry Dolstra
- Subjects
CD86 ,T cell ,Immunology ,hemic and immune systems ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Haematopoiesis ,medicine.anatomical_structure ,Immune system ,Antigen ,medicine ,Cancer research ,Ex vivo ,CD8 ,CD80 - Abstract
The prominent role of dendritic cells (DCs) in T cell activation is the rational for DC-based immunotherapy of cancer and infectious diseases. In cancer, DC therapy aims to induce tumor-specific effector T cell responses that can reduce or eliminate the tumor, and to develop immunological memory to control tumor relapse. So far, the vast majority of DC vaccination studies have been performed with DCs differentiated from monocytes (Mo-DCs) that are loaded with tumor-associated antigens (TAAs) or minor histocompatibility antigens (MiHA). This strategy has been reported to induce the expansion of antigen-specific CD4+ and/or CD8+ T cells in the majority of patients, however only a fraction of the patients develop clinical responses. Strategies to improve the potency of DC-based vaccines are to increase the stimulatory and migratory capacity of Mo-DCs, or to use alternative DC subtypes, such as naturally circulating plasmacytoid DCs (pDCs), BDCA1+ myeloid DCs (mDCs) or BDCA3+ mDCs. These DC subsets are potent inducers of antigen-specific T cell responses, and are therefore attractive cells to exploit for DC-based therapy. However, since their frequency in blood is very low, it is a challenge to obtain high enough numbers for immunotherapy. It would be advantageous if DCs, which are phenotypically and functionally similar to blood pDCs and mDCs, could be generated from CD34+ hematopoietic progenitor cells (HPCs). Interestingly, recent findings have indicated that the aryl hydrocarbon receptor (AhR) not only regulates toxic effects of environmental contaminants, but also plays a role in modulating hematopoiesis and the immune system. For instance, it has been reported that StemRegenin 1 (SR1), a small molecule inhibitor of AhR, promotes the ex vivo expansion of human CD34+ HPCs that are able to effectively engraft immunodeficient mice. Furthermore, differentiation of Langerhans cells and monocytes in vitro from HPCs can be inhibited by the addition of the AhR agonist VAF347. In light of these data, we investigated if we could generate DC subsets from CD34+ HPCs by supplementing SR1. Therefore, we cultured CD34+ HPCs in medium containing SCF, Flt3L, IL-6, TPO supplemented with 1 μM SR1 or DMSO as control. Interestingly, addition of SR1 explicitly promoted the emergence of pDCs (CD11c-HLA-DR+CD123hiBDCA2+BDCA4+ cells), BDCA1+ mDCs (Lin1-HLA-DR+BDCA1+BDCA3- cells) and BDCA3+ mDCs (Lin1-HLA-DR+BDCA1-BDCA3+ cells). After three weeks of culture, the frequency of these DC subsets was significantly higher in cultures with SR1 compared to control conditions; 2.9% vs. 0.04% for pDCs, 4.6% vs. 0.5% for BDCA1+ mDCs and 1.1% vs. 0.1% for BDCA3+ mDCs (n=3-5 donors). The average yield after three weeks of culture with SR1 starting from 105 CD34+ UCB cells was 3.8x106 pDCs, 5.3x106 BDCA1+ mDCs and 1.2x106 BDCA3+ mDCs (n=3-5 donors). Furthermore, SR1 also promoted the differentiation of DC subsets from CD34+ cells obtained from peripheral blood of G-CSF-mobilized donors. The average frequency of DCs in these SR1-cultures was 4.7%, 3.8% and 0.9% for pDCs, BDCA1+ and BDCA3+ mDCs, respectively (n=3 donors), which is comparable to the frequency obtained from UCB CD34+ cells. But the expansion potential of G-CSF-mobilized blood CD34+ HPCs was lower than that of UCB CD34+ cells, resulting in average DC yields of 0.6x106, 0.5x106 and 0.1x106 from 105 CD34+ cells (n=3). Flow cytometry analysis demonstrated that the SR1-induced pDCs and mDCs are phenotypically comparable to their naturally occurring counterpart in blood. Furthermore, the ex vivo-generated pDCs potently responded to stimulation with TLR7 and TLR9 ligands by secreting high amounts of IFN-α and upregulating CD83, CD80, CD86 and CCR7. The HPC-mDC subsets also upregulate CD80 and CD83 upon TLR3, TLR4 or TLR7/8 ligation. Finally, both the ex vivo-generated pDCs and mDCs induced potent allogeneic T cell responses and activated CD8+ effector T cells against hematopoietic-restricted MiHA. These findings demonstrate that our SR1 culture system not only allows detailed study of DC differentiation and molecular regulations in vitro, but it also offers the opportunity to evaluate the in vivo efficacy of cultured DC subsets upon vaccination into patients with cancer and viral infections. Disclosures: Spanholtz: Glycostem Therapeutics: Employment.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.