19 results on '"Robbert Van Der Voort"'
Search Results
2. CXCR4, but not CXCR3, drives CD8(+) T-cell entry into and migration through the murine bone marrow
- Author
-
Graham Anderson, Mark Hoogenboezem, Edith Slot, Martijn A. Nolte, Marieke Goedhart, Stephanie Gessel, Carlijn Voermans, Jaap D. van Buul, Ellis Gielen, Sulima Geerman, Beth Lucas, Stephan Huveneers, Robbert van der Voort, Timo Rademakers, Harry Dolstra, Graduate School, AII - Inflammatory diseases, Landsteiner Laboratory, Medical Biochemistry, ACS - Atherosclerosis & ischemic syndromes, ACS - Microcirculation, Clinical Haematology, and CCA - Cancer biology and immunology
- Subjects
0301 basic medicine ,EXPRESSION ,Stromal cell ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Immunology ,T cells ,EFFECTOR ,CHEMOKINE RECEPTOR CXCR3 ,Biology ,CXCR3 ,LYMPHOCYTES ,CXCR4 ,03 medical and health sciences ,Chemokine receptor ,All institutes and research themes of the Radboud University Medical Center ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Bone marrow ,HEMATOPOIETIC STEM-CELLS ,Migration ,IN-VIVO ,stromal cells ,MEMORY ,PROLIFERATION ,CXCL12 ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,MAINTENANCE ,SURVIVAL ,CD8 ,030215 immunology ,Homing (hematopoietic) - Abstract
The BM serves as a blood-forming organ, but also supports the maintenance and immune surveillance function of many T cells. Yet, in contrast to other organs, little is known about the molecular mechanisms that drive T-cell migration to and localization inside the BM. As BM accumulates many CXCR3-expressing memory CD8+ T cells, we tested the involvement of this chemokine receptor, but found that CXCR3 is not required for BM entry. In contrast, we could demonstrate that CXCR4, which is highly expressed on both naive and memory CD8+ T cells in BM, is critically important for homing of all CD8+ T-cell subsets to the BM in mice. Upon entry into the BM parenchyma, both naïve and memory CD8+ T cells locate close to sinusoidal vessels. Intravital imaging experiments revealed that CD8 T cells are surprisingly immobile and we found that they interact with ICAM-1+VCAM-1+BP-1+ perivascular stromal cells. These cells are the major source of CXCL12, but also express key survival factors and maintenance cytokines IL-7 and IL-15. We therefore conclude that CXCR4 is not only crucial for entry of CD8+ T cells into the BM, but also controls their subsequent localization toward BM niches that support their survival.
- Published
- 2019
3. Inhibition of Akt signaling promotes the generation of superior tumor-reactive T cells for adoptive immunotherapy
- Author
-
Harry Dolstra, Frans Maas, J.H. Frederik Falkenburg, Cynthia S. M. Kramer, Michel G.D. Kester, Nicolaas Schaap, Willemijn Hobo, Noortje M. P. van de Weem, Luca Gattinoni, Robbert van der Voort, Anniek B. van der Waart, and Joop H. Jansen
- Subjects
Cytotoxicity, Immunologic ,Transplantation Conditioning ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,medicine.medical_treatment ,Immunology ,Priming (immunology) ,Mice, Transgenic ,Mice, SCID ,CD8-Positive T-Lymphocytes ,Immunotherapy, Adoptive ,Biochemistry ,Minor Histocompatibility Antigens ,Mice ,Mice, Inbred NOD ,Cancer stem cell ,Quinoxalines ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Protein Kinase Inhibitors ,Cells, Cultured ,Transplantation ,business.industry ,CD28 ,Cell Biology ,Hematology ,Immunotherapy ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Cell biology ,Benzimidazoles ,Female ,Stem cell ,business ,Proto-Oncogene Proteins c-akt ,CD8 ,Ex vivo ,Signal Transduction - Abstract
Item does not contain fulltext Effective T-cell therapy against cancer is dependent on the formation of long-lived, stem cell-like T cells with the ability to self-renew and differentiate into potent effector cells. Here, we investigated the in vivo existence of stem cell-like antigen-specific T cells in allogeneic stem cell transplantation (allo-SCT) patients and their ex vivo generation for additive treatment posttransplant. Early after allo-SCT, CD8(+) stem cell memory T cells targeting minor histocompatibility antigens (MiHAs) expressed by recipient tumor cells were not detectable, emphasizing the need for improved additive MiHA-specific T-cell therapy. Importantly, MiHA-specific CD8(+) T cells with an early CCR7(+)CD62L(+)CD45RO(+)CD27(+)CD28(+)CD95(+) memory-like phenotype and gene signature could be expanded from naive precursors by inhibiting Akt signaling during ex vivo priming and expansion. This resulted in a MiHA-specific CD8(+) T-cell population containing a high proportion of stem cell-like T cells compared with terminal differentiated effector T cells in control cultures. Importantly, these Akt-inhibited MiHA-specific CD8(+) T cells showed a superior expansion capacity in vitro and in immunodeficient mice and induced a superior antitumor effect in intrafemural multiple myeloma-bearing mice. These findings provide a rationale for clinical exploitation of ex vivo-generated Akt-inhibited MiHA-specific CD8(+) T cells in additive immunotherapy to prevent or treat relapse in allo-SCT patients.
- Published
- 2014
4. 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
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. Decreased Levels of Circulating IL17-Producing CD161(+)CCR6(+) T Cells Are Associated with Graft-versus-Host Disease after Allogeneic Stem Cell Transplantation
- Author
-
Nicole M. A. Blijlevens, Astrid G. S. van Halteren, Anniek B. van der Waart, M. Leenders, Walter J.F.M. van der Velden, Ton Feuth, Harry Dolstra, and Robbert van der Voort
- Subjects
CD4-Positive T-Lymphocytes ,Male ,T-Lymphocytes ,Graft vs Host Disease ,lcsh:Medicine ,CD8-Positive T-Lymphocytes ,Adaptive Immunity ,Interleukin 21 ,Cell Movement ,immune system diseases ,Molecular Cell Biology ,Cytotoxic T cell ,Bone Marrow and Stem Cell Transplantation ,lcsh:Science ,Immune Regulation Translational research [NCMLS 2] ,Multidisciplinary ,T Cells ,Histocompatibility Testing ,Stem Cells ,Interleukin-17 ,hemic and immune systems ,Hematology ,Middle Aged ,Phenotype ,medicine.anatomical_structure ,surgical procedures, operative ,Cyclosporine ,Medicine ,Female ,Immunotherapy ,Interleukin 17 ,Cellular Types ,Stem cell ,NK Cell Lectin-Like Receptor Subfamily B ,Research Article ,Adult ,Receptors, CCR6 ,Tumor Immunology ,Invasive mycoses and compromised host Translational research [N4i 2] ,Immune Cells ,T cell ,Immunology ,chemical and pharmacologic phenomena ,Biology ,Interferon-gamma ,Young Adult ,Immune system ,medicine ,Humans ,Transplantation, Homologous ,Transplantation ,Chemokine CCL20 ,lcsh:R ,Immunity ,Immunologic Subspecialties ,medicine.disease ,Graft-versus-host disease ,Evaluation of complex medical interventions [NCEBP 2] ,Case-Control Studies ,Multivariate Analysis ,lcsh:Q ,Immunologic Memory ,Stem Cell Transplantation - Abstract
Contains fulltext : 110799.pdf (Publisher’s version ) (Open Access) The C-type lectin-like receptor CD161 is a well-established marker for human IL17-producing T cells, which have been implicated to contribute to the development of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (allo-SCT). In this study, we analyzed CD161(+) T cell recovery, their functional properties and association with GVHD occurrence in allo-SCT recipients. While CD161(+)CD4(+) T cells steadily recovered, CD161(hi)CD8(+) T cell numbers declined during tapering of Cyclosporine A (CsA), which can be explained by their initial growth advantage over CD161(neg/low)CD8(+) T cells due to ABCB1-mediated CsA efflux. Interestingly, occurrence of acute and chronic GVHD was significantly correlated with decreased levels of circulating CD161(+)CD4(+) as well as CD161(hi)CD8(+) T cells. In addition, these subsets from transplanted patients secreted high levels of IFNgamma and IL17. Moreover, we found that CCR6 co-expression by CD161(+) T cells mediated specific migration towards CCL20, which was expressed in GVHD biopsies. Finally, we demonstrated that CCR6(+) T cells indeed were present in these CCL20(+) GVHD-affected tissues. In conclusion, we showed that functional CD161(+)CCR6(+) co-expressing T cells disappear from the circulation and home to GVHD-affected tissue sites. These findings support the hypothesis that CCR6(+)CD161-expressing T cells may be involved in the immune pathology of GVHD following their CCL20-dependent recruitment into affected tissues.
- Published
- 2012
8. Coinhibitory molecules in hematologic malignancies: targets for therapeutic intervention
- Author
-
Robbert van der Voort, Harry Dolstra, Wieger J. Norde, and Willemijn Hobo
- Subjects
Tumor microenvironment ,Immune Regulation Translational research [NCMLS 2] ,Stromal cell ,LAG3 ,business.industry ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Acquired immune system ,Biochemistry ,Immune system ,Hematologic Neoplasms ,Cancer cell ,Immune Tolerance ,Cytotoxic T cell ,Medicine ,Humans ,business - Abstract
The adaptive immune system can be a potent defense mechanism against cancer; however, it is often hampered by immune suppressive mechanisms in the tumor microenvironment. Coinhibitory molecules expressed by tumor cells, immune cells, and stromal cells in the tumor milieu can dominantly attenuate T-cell responses against cancer cells. Today, a variety of coinhibitory molecules, including cytotoxic T lymphocyte–associated antigen-4, programmed death-1, B and T lymphocyte attenuator, LAG3, T-cell immunoglobulin and mucin domain 3, and CD200 receptor, have been implicated in immune escape of cancer cells. Sustained signaling via these coinhibitory molecules results in functional exhaustion of T cells, during which the ability to proliferate, secrete cytokines, and mediate lysis of tumor cells is sequentially lost. In this review, we discuss the influence of coinhibitory pathways in suppressing autologous and allogeneic T cell–mediated immunity against hematologic malignancies. In addition, promising preclinical and clinical data of immunotherapeutic approaches interfering with negative cosignaling, either as monotherapy or in conjunction with vaccination strategies, are reviewed. Numerous studies indicate that coinhibitory signaling hampers the clinical benefit of current immunotherapies. Therefore, manipulation of coinhibitory networks is an attractive adjuvant immunotherapeutic intervention for hematologic cancers after standard treatment with chemotherapy and hematopoietic stem cell transplantation.
- Published
- 2012
9. B and T lymphocyte attenuator mediates inhibition of tumor-reactive CD8+ T cells in patients after allogeneic stem cell transplantation
- Author
-
Karen Schellens, Alan J. Korman, J.H. Frederik Falkenburg, Robbert van der Voort, Frans Maas, Hanny Fredrix, Harry Dolstra, Wieger J. Norde, Nicolaas Schaap, Willemijn Hobo, and Daniel Olive
- Subjects
T cell ,Immunology ,Epitopes, T-Lymphocyte ,BTLA ,CD8-Positive T-Lymphocytes ,Biology ,Minor Histocompatibility Antigens ,Interleukin 21 ,Translational research [ONCOL 3] ,Cell Line, Tumor ,Tumor Cells, Cultured ,medicine ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Receptors, Immunologic ,Antibodies, Blocking ,Antigen-presenting cell ,Immune Regulation Translational research [NCMLS 2] ,ZAP70 ,Hematopoietic Stem Cell Transplantation ,Natural killer T cell ,medicine.anatomical_structure ,Gene Targeting ,Neoplasm Recurrence, Local ,Immunologic Memory ,Receptors, Tumor Necrosis Factor, Member 14 - Abstract
Allogeneic stem cell transplantation (allo-SCT) can cure hematological malignancies by inducing alloreactive T cell responses targeting minor histocompatibility antigens (MiHA) expressed on malignant cells. Despite induction of robust MiHA-specific T cell responses and long-term persistence of alloreactive memory T cells specific for the tumor, often these T cells fail to respond efficiently to tumor relapse. Previously, we demonstrated the involvement of the coinhibitory receptor programmed death-1 (PD-1) in suppressing MiHA-specific CD8+ T cell immunity. In this study, we investigated whether B and T lymphocyte attenuator (BTLA) plays a similar role in functional impairment of MiHA-specific T cells after allo-SCT. In addition to PD-1, we observed higher BTLA expression on MiHA-specific CD8+ T cells compared with that of the total population of CD8+ effector-memory T cells. In addition, BTLA’s ligand, herpes virus entry mediator (HVEM), was found constitutively expressed by myeloid leukemia, B cell lymphoma, and multiple myeloma cells. Interference with the BTLA–HVEM pathway, using a BTLA blocking Ab, augmented proliferation of BTLA+PD-1+ MiHA-specific CD8+ T cells by HVEM-expressing dendritic cells. Notably, we demonstrated that blocking of BTLA or PD-1 enhanced ex vivo proliferation of MiHA-specific CD8+ T cells in respectively 7 and 9 of 11 allo-SCT patients. Notably, in 3 of 11 patients, the effect of BTLA blockade was more prominent than that of PD-1 blockade. Furthermore, these expanded MiHA-specific CD8+ T cells competently produced effector cytokines and degranulated upon Ag reencounter. Together, these results demonstrate that BTLA–HVEM interactions impair MiHA-specific T cell functionality, providing a rationale for interfering with BTLA signaling in post-stem cell transplantation therapies.
- Published
- 2012
10. 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
11. 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
12. 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
13. Myeloid leukemic progenitor cells can be specifically targeted by minor histocompatibility antigen LRH-1-reactive cytotoxic T cells
- Author
-
Inge Jedema, Ingrid M. Overes, J.H. Frederik Falkenburg, Johanna C. M. Vos, Robbert van der Voort, Hanny Fredrix, Michel G.D. Kester, Harry Dolstra, Anton Schattenberg, Theo de Witte, Frans Maas, and Wieger J. Norde
- Subjects
Adult ,Male ,Myeloid ,Immunology ,Gene Expression ,Graft vs Host Disease ,Antigens, CD34 ,X-Linked Inhibitor of Apoptosis Protein ,Biology ,Biochemistry ,Immune Regulation [NCMLS 2] ,Translational research [ONCOL 3] ,hemic and lymphatic diseases ,Minor histocompatibility antigen ,medicine ,Humans ,Cytotoxic T cell ,RNA, Messenger ,Progenitor cell ,Interleukin 3 ,Receptors, Purinergic P2 ,Reverse Transcriptase Polymerase Chain Reaction ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,Cell Biology ,Hematology ,Middle Aged ,Flow Cytometry ,medicine.disease ,DNA-Binding Proteins ,Leukemia ,medicine.anatomical_structure ,Leukemia, Myeloid ,Neoplastic Stem Cells ,Female ,Receptors, Purinergic P2X5 ,CD8 ,T-Lymphocytes, Cytotoxic ,Transcription Factors - Abstract
Contains fulltext : 79829.pdf (Publisher’s version ) (Closed access) CD8(+) T cells recognizing minor histocompatibility antigens (MiHAs) on leukemic stem and progenitor cells play a pivotal role in effective graft-versus-leukemia reactivity after allogeneic stem cell transplantation (SCT). Previously, we identified a hematopoiesis-restricted MiHA, designated LRH-1, which is presented by HLA-B7 and encoded by the P2X5 purinergic receptor gene. We found that P2X5 is significantly expressed in CD34(+) leukemic subpopulations from chronic myeloid leukemia (CML) and acute myeloid leukemia (AML) patients. Here, we demonstrate that LRH-1-specific CD8(+) T-cell responses are frequently induced in myeloid leukemia patients following donor lymphocyte infusions. Patients with high percentages of circulating LRH-1-specific CD8(+) T cells had no or only mild graft-versus-host disease. Functional analysis showed that LRH-1-specific cytotoxic T lymphocytes (CTLs) isolated from 2 different patients efficiently target LRH-1-positive leukemic CD34(+) progenitor cells from both CML and AML patients, whereas mature CML cells are only marginally lysed due to down-regulation of P2X5. Furthermore, we observed that relative resistance to LRH-1 CTL-mediated cell death due to elevated levels of antiapoptotic XIAP could be overcome by IFN-gamma prestimulation and increased CTL-target ratios. These findings provide a rationale for use of LRH-1 as immunotherapeutic target antigen to treat residual or persisting myeloid malignancies after allogeneic SCT.
- Published
- 2009
14. Expression of P2X5 in lymphoid malignancies results in LRH-1-specific cytotoxic T-cell-mediated lysis
- Author
-
Harry Dolstra, J Han J M van Krieken, Joop H. Jansen, Ingrid M. Overes, Björn de Rijke, Theo de Witte, Robbert van der Voort, Reinier Raymakers, Agnes van Horssen-Zoetbrood, Hanny Fredrix, and Aniek O. de Graaf
- Subjects
Cytotoxicity, Immunologic ,Age-related aspects of cancer [ONCOL 2] ,Genetics and epigenetic pathways of disease [NCMLS 6] ,medicine.medical_treatment ,Biology ,Donor lymphocyte infusion ,Immune Regulation [NCMLS 2] ,Translational research [ONCOL 3] ,medicine ,Minor histocompatibility antigen ,Tumor Cells, Cultured ,Iron metabolism [IGMD 7] ,Cytotoxic T cell ,Humans ,RNA, Messenger ,RNA, Neoplasm ,Molecular diagnosis, prognosis and monitoring [UMCN 1.2] ,Hereditary cancer and cancer-related syndromes [ONCOL 1] ,Receptors, Purinergic P2 ,Reverse Transcriptase Polymerase Chain Reaction ,Lymphoma, Non-Hodgkin ,Immunotherapy, gene therapy and transplantation [UMCN 1.4] ,Hematology ,Immunotherapy ,medicine.disease ,Lymphoma ,Leukemia, Lymphoid ,Neoplasm Proteins ,Tumor microenvironment [UMCN 1.3] ,DNA-Binding Proteins ,CTL ,Haematopoiesis ,Immunology ,Stem cell ,Multiple Myeloma ,Receptors, Purinergic P2X5 ,Immunity, infection and tissue repair [NCMLS 1] ,T-Lymphocytes, Cytotoxic ,Transcription Factors - Abstract
Contains fulltext : 70311.pdf (Publisher’s version ) (Closed access) Minor histocompatibility antigens (MiHA) selectively expressed by haematopoietic cells are attractive targets for specific immunotherapy after allogeneic stem cell transplantation (SCT). Previously, we described LRH-1 as a haematopoietic-lineage restricted MiHA that is capable of eliciting an allogeneic cytotoxic T-lymphocyte (CTL) response after SCT and donor lymphocyte infusion. Importantly, the gene encoding LRH-1, P2X5, is not expressed in prominent graft-versus-host-disease target tissues such as skin, liver and gut. Here, we investigate whether LRH-1-specific immunotherapy may be exploited for the treatment of lymphoid malignancies. We examined P2X5 mRNA expression in a large panel of patient samples and cell lines from different types of lymphoid malignancies by real-time quantitative reverse transcription polymerase chain reaction. P2X5 mRNA was highly expressed in malignant cells from all stages of lymphoid development. Furthermore, all LRH-1-positive lymphoid tumour cell lines were susceptible to LRH-1 CTL-mediated lysis in flow cytometry-based cytotoxicity assays. However, interferon-gamma production was low or absent after stimulation with some cell lines, possibly due to differences in activation thresholds for CTL effector functions. Importantly, primary cells from patients with lymphoid malignancies were effectively lysed by LRH-1-specific CTL. These findings indicate that MiHA LRH-1 is a potential therapeutic target for cellular immunotherapy of lymphoid malignancies.
- Published
- 2008
15. 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
16. 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
17. CXCR3 Is Required for the Efficient Recruitment of Bone Marrow-Resident Memory T Cells to Inflamed Tissues
- Author
-
Bao Lu, Thomas J. H. Volman, Martijn A. Nolte, Claudia Brandao, Harry Dolstra, Vivienne Verweij, Robbert van der Voort, Craig Gerard, and Klaas P. J. M. van Gisbergen
- Subjects
T cell ,Immunology ,Cell Biology ,Hematology ,Biology ,Natural killer T cell ,Biochemistry ,Molecular biology ,CCL5 ,Interleukin 21 ,medicine.anatomical_structure ,medicine ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Memory T cell - Abstract
Abstract 3242 Although the importance of the bone marrow (BM) in hematopoiesis is well known, its function in adaptive immune responses has only recently been acknowledged. Currently it is known that the BM contains fully functional CD4+ and CD8+ T cells that can engage in both primary and secondary immune responses. Interestingly, most of these T cells belong to the memory T cell lineage, identifying the BM as one of the largest memory T cell reservoirs in the body. Since not much is known about the trafficking of BM T cells, we compared the homing phenotype and function of T cell subsets in the BM, blood, spleen and peripheral lymph nodes (pLN). In addition, we determined the expression of chemokine mRNA and protein levels in the BM and other lymphoid organs. We confirmed that at least 80% of the CD4+ and 60% of the CD8+ BM T cells have a memory phenotype, and that most CD4+ T cells belong to the effector memory lineage, while the CD8+ population predominantly consists of central memory T cells. Most BM T cells expressed the chemokine receptor CXCR3, the adhesion molecules P-selectin glycoprotein ligand 1 and VLA-4, and increased levels of CD44 and LFA-1, as compared to T cells from the spleen. In addition, L-selectin was absent from most CD4+ BM T cells, but present on virtually all CD8+ T cells. Notably, the percentage of CXCR3+ T cells within the effector memory and central memory subsets from BM was higher than within the same subsets from pLN. Furthermore, BM contained significant mRNA levels of the CXCR3 ligands CXCL9, CXCL10 and CXCL11. An in vivo migration assay using a mixture of fluorescent-labeled T cells from CXCR3-deficient mice and control mice indicated however that during homeostasis CXCR3 does not play a major role in BM entry or retention. These data suggest that CXCR3 expressed by memory T cells is rather involved in BM exit, than in BM entry. Indeed, we observed that, as compared to control mice, CXCR3−/− mice contained significantly more CD4+ and CD8+ T cells in their BM. Additional in vitro assays demonstrated that CD4+ and CD8+ BM T cells migrated vigorously in response to CXCL9 and CXCL10, generally released in high concentrations during inflammation. Finally, we demonstrate that CXCR3−/− effector/effector memory T cells, but not wild type T cells, accumulate in the BM of mice infected with lymphocytic choriomeningitis virus. Altogether, these data demonstrate that the BM is a major reservoir of memory T cells that employ CXCR3 to quickly respond to chemotactic signals from inflamed tissues. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2011
- Full Text
- View/download PDF
18. Use of RNA Electroporated DC for Activation of LRH-1 Specific Cytotoxic T Lymphocytes in the Treatment of Lymphoid Malignancies
- Author
-
Han van Krieken, Reinier Raymakers, Hanny Fredrix, Robbert van der Voort, Ingrid M. Overes, Agnes van Horssen-Zoetbrood, Harry Dolstra, T. de Witte, and Björn de Rijke
- Subjects
medicine.medical_treatment ,T cell ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Immunotherapy ,Biology ,medicine.disease ,Biochemistry ,CTL ,medicine.anatomical_structure ,Graft-versus-host disease ,Antigen ,medicine ,Minor histocompatibility antigen ,Cytotoxic T cell - Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potent treatment for patients with hematological malignancies. The therapeutic efficacy 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. Unfortunately, GVT response is often accompanied by graft-versus-host disease (GVHD) causing severe damage to skin, liver and gut. Therefore, it would be highly beneficial to direct GVT immunity to MiHA that are selectively expressed by “malignant” hematopoietic cells. Recently, we identified a novel lymphoid lineage-restricted MiHA, designated LRH-1, which is derived from a frame shift polymorphism in the P2X5 purinergic receptor protein (J. Clin. Invest.2005:115:3506–3516). Here, we examined by real-time quantitative RT-PCR mRNA expression of P2X5 in patient samples and cell lines from numerous lymphoid malignancies. We observed that P2X5 mRNA is highly expressed in tumor cells from all stages of lymphoid development. Furthermore, we demonstrated that LRH-1−specific CTL efficiently lyse LRH-1+ tumor cells and cell lines of lymphoid origin. These findings illustrate that LRH-1 is an attractive target for specific immunotherapy after allogeneic HSCT. A potentially efficient strategy is the application of MiHA-loaded dendritic cells (DC) to boost MiHA-specific T cell responses already primed in vivo after HSCT. To facilitate efficient presentation of LRH-1 by DC, we have optimized RNA electroporation of DC using in vitro-transcribed mRNA. We observed that RNA electroporation of mature DC resulted in a high yield of viable cells, high expression of co-stimulatory molecules, no loss of migratory capacity towards lymph node-specific chemokines, and high protein expression of the introduced antigens. Furthermore, we demonstrated that RNA-electroporated DC display long-lasting peptide presentation to LRH-1− specific CTL and induce proliferation of LRH-1− specific effector-memory CTL ex vivo. These data will be of importance in designing LRH-1− based immunotherapy in transplanted patients with lymphoid malignancies.
- Published
- 2006
- Full Text
- View/download PDF
19. 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
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.