36 results on '"van der Waart, Anniek B."'
Search Results
2. Increased peritoneal TGF-b1 is associated with ascites-induced NK-cell dysfunction and reduced survival in high-grade epithelial ovarian cancer.
- Author
-
Maas, Ralph J. A., Hoogstad-van Evert, Janneke S., Hagemans, Iris M., Brummelman, Jolanda, van Ens, Diede, de Jonge, Paul K. J. D., Hooijmaijers, Laura, Mahajan, Shweta, van der Waart, Anniek B., Hermans, Charlotte K. J. C., de Klein, Janne, Woestenenk, Rob, van Herwaarden, Antonius E., Schaap, Nicolaas P. M., Rezaeifard, Somayeh, Tauriello, Daniele V. F., Zusterzeel, Petra L. M., Ottevanger, Nelleke, Jansen, Joop H., and Hobo, Willemijn
- Subjects
KILLER cells ,REGULATORY T cells ,OVARIAN epithelial cancer ,CELL anatomy ,TRANSFORMING growth factors - Abstract
Natural killer (NK) cell therapy represents an attractive immunotherapy approach against recurrent epithelial ovarian cancer (EOC), as EOC is sensitive to NK cellmediated cytotoxicity. However, NK cell antitumor activity is dampened by suppressive factors in EOC patient ascites. Here, we integrated functional assays, soluble factor analysis, high-dimensional flow cytometry cellular component data and clinical parameters of advanced EOC patients to study the mechanisms of ascites-induced inhibition of NK cells. Using a suppression assay, we found that ascites from EOC patients strongly inhibits peripheral blood-derived NK cells and CD34+ progenitor-derived NK cells, albeit the latter were more resistant. Interestingly, we found that higher ascites-induced NK cell inhibition correlated with reduced progression-free and overall survival in EOC patients. Furthermore, we identified transforming growth factor (TGF)-b1 to correlate with ascites-induced NK cell dysfunction and reduced patient survival. In functional assays, we showed that proliferation and anti-tumor reactivity of CD34+ progenitor-derived NK cells are significantly affected by TGF-b1 exposure. Moreover, inhibition of TGF-b1 signaling with galunisertib partly restored NK cell functionality in some donors. For the cellular components, we showed that the secretome is associated with a different composition of CD45+ cells between ascites of EOC and benign reference samples with higher proportions of macrophages in the EOC patient samples. Furthermore, we revealed that higher TGF-b1 levels are associated with the presence of M2-like macrophages, B cell populations and T-regulatory cells in EOC patient ascites. These findings reveal that targeting TGF-b1 signaling could increase NK cell immune responses in high-grade EOC patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Cell composition and expansion strategy can reduce the beneficial effect of AKT-inhibition on functionality of CD8+ T cells
- Author
-
Mousset, Charlotte M., Hobo, Willemijn, de Ligt, Aafke, Baardman, Sjoerd, Schaap, Nicolaas P. M., Jansen, Joop H., van der Waart, Anniek B., and Dolstra, Harry
- Published
- 2020
- Full Text
- View/download PDF
4. AKT inhibition generates potent polyfunctional clinical grade AUTO1 CAR T-cells, enhancing function and survival
- Author
-
Mehra, Vedika, primary, Agliardi, Giulia, additional, Dias Alves Pinto, Juliana, additional, Shafat, Manar S, additional, Garai, Amaia Cadinanos, additional, Green, Louisa, additional, Hotblack, Alastair, additional, Arce Vargas, Fred, additional, Peggs, Karl S, additional, van der Waart, Anniek B, additional, Dolstra, Harry, additional, Pule, Martin A, additional, and Roddie, Claire, additional
- Published
- 2023
- Full Text
- View/download PDF
5. Engineering of CD34+ progenitor-derived natural killer cells with higher-affinity CD16a for enhanced antibody-dependent cellular cytotoxicity
- Author
-
van Hauten, Paulien M.M., Hooijmaijers, Laura, Vidal-Manrique, Marcos, van der Waart, Anniek B., Hobo, Willemijn, Wu, Jianming, Blijlevens, Nicole M.A., Jansen, Joop H., Walcheck, Bruce, Schaap, Nicolaas P.M., de Jonge, Paul K.J.D., and Dolstra, Harry
- Published
- 2024
- Full Text
- View/download PDF
6. Human CD34+-derived complete plasmacytoid and conventional dendritic cell vaccine effectively induces antigen-specific CD8+ T cell and NK cell responses in vitro and in vivo.
- Author
-
van Eck van der Sluijs, Jesper, van Ens, Diede, Brummelman, Jolanda, Heister, Daan, Sareen, Aastha, Truijen, Lisa, van Ingen Schenau, Dorette S., Heemskerk, Mirjam H. M., Griffioen, Marieke, Kester, Michel G. D., Schaap, Nicolaas P. M., Jansen, Joop H., van der Waart, Anniek B., Dolstra, Harry, and Hobo, Willemijn
- Abstract
Allogeneic stem cell transplantation (alloSCT) can be curative for hemato-oncology patients due to effective graft-versus-tumor immunity. However, relapse remains the major cause of treatment failure, emphasizing the need for adjuvant immunotherapies. In this regard, post-transplantation dendritic cell (DC) vaccination is a highly interesting strategy to boost graft-versus-tumor responses. Previously, we developed a clinically applicable protocol for simultaneous large-scale generation of end-stage blood DC subsets from donor-derived CD34
+ stem cells, including conventional type 1 and 2 DCs (cDC1s and cDC2s), and plasmacytoid DCs (pDCs). In addition, the total cultured end-product (DC-complete vaccine), also contains non-end-stage-DCs (i.e. non-DCs). In this study, we aimed to dissect the phenotypic identity of these non-DCs and their potential immune modulatory functions on the potency of cDCs and pDCs in stimulating tumor-reactive CD8+ T and NK cell responses, in order to obtain rationale for clinical translation of our DC-complete vaccine. The non-DC compartment was heterogeneous and comprised of myeloid progenitors and (immature) granulocyte- and monocyte-like cells. Importantly, non-DCs potentiated toll-like receptor-induced DC maturation, as reflected by increased expression of co-stimulatory molecules and enhanced cDC-derived IL-12 and pDC-derived IFN-α production. Additionally, antigen-specific CD8+ T cells effectively expanded upon DC-complete vaccination in vitro and in vivo. This effect was strongly augmented by non-DCs in an antigen-independent manner. Moreover, non-DCs did not impair in vitro DC-mediated NK cell activation, degranulation nor cytotoxicity. Notably, in vivo i.p. DC-complete vaccination activated i.v. injected NK cells. Together, these data demonstrate that the non-DC compartment potentiates DC-mediated activation and expansion of antigen-specific CD8+ T cells and do not impair NK cell responses in vitro and in vivo. This underscores the rationale for further clinical translation of our CD34+ -derived DC-complete vaccine in hemato-oncology patients post alloSCT. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
7. Inhibition of Akt signaling promotes the generation of superior tumor-reactive T cells for adoptive immunotherapy
- Author
-
van der Waart, Anniek B., Noortje van de Weem, M.P., Maas, Frans, Kramer, Cynthia S.M., Michel Kester, G.D., Frederik Falkenburg, J.H., Schaap, Nicolaas, Jansen, Joop H., van der Voort, Robbert, Gattinoni, Luca, Hobo, Willemijn, and Dolstra, Harry
- Published
- 2014
- Full Text
- View/download PDF
8. Targeting the IL17 Pathway for the Prevention of Graft-Versus-Host Disease
- Author
-
van der Waart, Anniek B., van der Velden, Walter J.F.M., Blijlevens, Nicole M., and Dolstra, Harry
- Published
- 2014
- Full Text
- View/download PDF
9. Clinically applicable CD34+-derived blood dendritic cell subsets exhibit key subset-specific features and potently boost anti-tumor T and NK cell responses
- Author
-
van Eck van der Sluijs, Jesper, van Ens, Diede, Thordardottir, Soley, Vodegel, Denise, Hermens, Inge, van der Waart, Anniek B., Falkenburg, J. H. Frederik, Kester, Michel G. D., de Rink, Iris, Heemskerk, Mirjam H. M., Borst, Jannie, Schaap, Nicolaas P. M., Jansen, Joop H., Xiao, Yanling, Dolstra, Harry, and Hobo, Willemijn
- Subjects
CD34+ hematopoietic progenitor cells ,Cancer Research ,T-Lymphocytes ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,medicine.medical_treatment ,Immunology ,Cell ,Cell Culture Techniques ,T cells ,CD34 ,Antigens, CD34 ,CD34(+) hematopoietic progenitor cells ,NK cells ,Biology ,Lymphocyte Activation ,Dendritic cells ,03 medical and health sciences ,Cross-Priming ,0302 clinical medicine ,Immunity ,medicine ,Humans ,Immunology and Allergy ,030304 developmental biology ,Antigen Presentation ,0303 health sciences ,Vaccination ,Dendritic cell ,Immunotherapy ,Hematopoietic Stem Cells ,Acquired immune system ,Clinical application ,Killer Cells, Natural ,Transplantation ,medicine.anatomical_structure ,Oncology ,Cancer research ,Original Article ,Stem cell ,030215 immunology - Abstract
Allogeneic stem cell transplantation (alloSCT), following induction chemotherapy, can be curative for hemato-oncology patients due to powerful graft-versus-tumor immunity. However, disease recurrence remains the major cause of treatment failure, emphasizing the need for potent adjuvant immunotherapy. In this regard, dendritic cell (DC) vaccination is highly attractive, as DCs are the key orchestrators of innate and adaptive immunity. Natural DC subsets are postulated to be more powerful compared with monocyte-derived DCs, due to their unique functional properties and cross-talk capacity. Yet, obtaining sufficient numbers of natural DCs, particularly type 1 conventional DCs (cDC1s), is challenging due to low frequencies in human blood. We developed a clinically applicable culture protocol using donor-derived G-CSF mobilized CD34+ hematopoietic progenitor cells (HPCs) for simultaneous generation of high numbers of cDC1s, cDC2s and plasmacytoid DCs (pDCs). Transcriptomic analyses demonstrated that these ex vivo-generated DCs highly resemble their in vivo blood counterparts. In more detail, we demonstrated that the CD141+CLEG9A+ cDC1 subset exhibited key features of in vivo cDC1s, reflected by high expression of co-stimulatory molecules and release of IL-12p70 and TNF-α. Furthermore, cDC1s efficiently primed alloreactive T cells, potently cross-presented long-peptides and boosted expansion of minor histocompatibility antigen-experienced T cells. Moreover, they strongly enhanced NK cell activation, degranulation and anti-leukemic reactivity. Together, we developed a robust culture protocol to generate highly functional blood DC subsets for in vivo application as tailored adjuvant immunotherapy to boost innate and adaptive anti-tumor immunity in alloSCT patients.
- Published
- 2021
- Full Text
- View/download PDF
10. 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
-
van der Waart, Anniek B., Fredrix, Hanny, van der Voort, Robbert, Schaap, Nicolaas, Hobo, Willemijn, and Dolstra, Harry
- Published
- 2015
- Full Text
- View/download PDF
11. Polymorphisms in CCR6 Are Associated with Chronic Graft-versus-Host Disease and Invasive Fungal Disease in Matched-Related Hematopoietic Stem Cell Transplantation
- Author
-
Broen, Kelly, van der Waart, Anniek B., Greupink-Draaisma, Annelies, Metzig, Julia, Feuth, Ton, Schaap, Nicolaas P.M., Blijlevens, Nicole M.A., van der Velden, Walter J.F.M., and Dolstra, Harry
- Published
- 2011
- Full Text
- View/download PDF
12. CD34+ progenitor-derived NK cell and gemcitabine combination therapy increases killing of ovarian cancer cells in NOD/SCID/IL2Rgnull mice
- Author
-
Van der Meer, Jolien M.R., primary, de Jonge, Paul K.J.D., additional, van der Waart, Anniek B., additional, Geerlings, Alexander C., additional, Moonen, Jurgen P., additional, Brummelman, Jolanda, additional, de Klein, Janne, additional, Vermeulen, Malou C., additional, Maas, Ralph J.A., additional, Schaap, Nicolaas P.M., additional, Hoogstad-van Evert, Janneke S., additional, Ottevanger, Petronella B., additional, Jansen, Joop H., additional, Hobo, Willemijn, additional, and Dolstra, Harry, additional
- Published
- 2021
- Full Text
- View/download PDF
13. Clinically applicable CD34+-derived blood dendritic cell subsets exhibit key subset-specific features and potently boost anti-tumor T and NK cell responses.
- Author
-
van Eck van der Sluijs, Jesper, van Ens, Diede, Thordardottir, Soley, Vodegel, Denise, Hermens, Inge, van der Waart, Anniek B., Falkenburg, J. H. Frederik, Kester, Michel G. D., de Rink, Iris, Heemskerk, Mirjam H. M., Borst, Jannie, Schaap, Nicolaas P. M., Jansen, Joop H., Xiao, Yanling, Dolstra, Harry, and Hobo, Willemijn
- Subjects
KILLER cells ,BLOOD cells ,DENDRITIC cells ,T cells ,PROGENITOR cells - Abstract
Allogeneic stem cell transplantation (alloSCT), following induction chemotherapy, can be curative for hemato-oncology patients due to powerful graft-versus-tumor immunity. However, disease recurrence remains the major cause of treatment failure, emphasizing the need for potent adjuvant immunotherapy. In this regard, dendritic cell (DC) vaccination is highly attractive, as DCs are the key orchestrators of innate and adaptive immunity. Natural DC subsets are postulated to be more powerful compared with monocyte-derived DCs, due to their unique functional properties and cross-talk capacity. Yet, obtaining sufficient numbers of natural DCs, particularly type 1 conventional DCs (cDC1s), is challenging due to low frequencies in human blood. We developed a clinically applicable culture protocol using donor-derived G-CSF mobilized CD34
+ hematopoietic progenitor cells (HPCs) for simultaneous generation of high numbers of cDC1s, cDC2s and plasmacytoid DCs (pDCs). Transcriptomic analyses demonstrated that these ex vivo-generated DCs highly resemble their in vivo blood counterparts. In more detail, we demonstrated that the CD141+ CLEG9A+ cDC1 subset exhibited key features of in vivo cDC1s, reflected by high expression of co-stimulatory molecules and release of IL-12p70 and TNF-α. Furthermore, cDC1s efficiently primed alloreactive T cells, potently cross-presented long-peptides and boosted expansion of minor histocompatibility antigen-experienced T cells. Moreover, they strongly enhanced NK cell activation, degranulation and anti-leukemic reactivity. Together, we developed a robust culture protocol to generate highly functional blood DC subsets for in vivo application as tailored adjuvant immunotherapy to boost innate and adaptive anti-tumor immunity in alloSCT patients. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
14. CD34+ progenitor-derived NK cell and gemcitabine combination therapy increases killing of ovarian cancer cells in NOD/SCID/IL2Rgnull mice.
- Author
-
Van der Meer, Jolien M. R., de Jonge, Paul K. J. D., van der Waart, Anniek B., Geerlings, Alexander C., Moonen, Jurgen P., Brummelman, Jolanda, de Klein, Janne, Vermeulen, Malou C., Maas, Ralph J. A., Schaap, Nicolaas P. M., Hoogstad-van Evert, Janneke S., Ottevanger, Petronella B., Jansen, Joop H., Hobo, Willemijn, and Dolstra, Harry
- Subjects
OVARIAN cancer ,KILLER cells ,CANCER cells ,GEMCITABINE ,DEATH receptors ,CELLULAR immunity - Abstract
Combining natural killer (NK) cell adoptive transfer with tumor-sensitizing chemotherapy is an attractive approach against recurrent ovarian cancer (OC), as OC is sensitive to NK cell-mediated immunity. Previously, we showed that CD34
+ hematopoietic progenitor cell (HPC)-derived NK cells can kill OC cells in vitro and inhibit OC tumor growth in mice. Here, we investigated the potential of HPC-NK cell therapy combined with chemotherapeutic gemcitabine (used in recurrent OC patients) against OC. We examined the phenotypical, functional, and cytotoxic effects of gemcitabine on HPC-NK cells and/or OC cells in vitro and in OC-bearing mice. To this end, we treated OC cells and/or HPC-NK cells with or without gemcitabine and analyzed the phenotype, cytokine production, and anti-tumor reactivity. We found that gemcitabine did not affect the phenotype and functionality of HPC-NK cells, while on OC cells expression of NK cell activating ligands and death receptors was upregulated. Although gemcitabine pre-treatment of OC cells did not improve the functionality of HPC-NK cells, importantly, HPC-NK cells and gemcitabine additively killed OC cells in vitro. Similarly, combined HPC-NK cell and gemcitabine treatment additively decreased tumor growth in OC-bearing mice. Collectively, our results indicate that combination therapy of HPC-NK cells and gemcitabine results in augmented OC killing in vitro and in vivo. This provides a rationale for exploring this therapeutic strategy in patients with recurrent OC. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
15. Cell composition and expansion strategy can reduce the beneficial effect of AKT-inhibition on functionality of CD8+ T cells.
- Author
-
Mousset, Charlotte M., Hobo, Willemijn, de Ligt, Aafke, Baardman, Sjoerd, Schaap, Nicolaas P. M., Jansen, Joop H., van der Waart, Anniek B., and Dolstra, Harry
- Subjects
T cells ,STEM cells ,CELLULAR therapy ,LYMPHOCYTES - Abstract
AKT-inhibition is a promising approach to improve T cell therapies; however, its effect on CD4
+ T cells is insufficiently explored. Previously, we and others showed that AKT-inhibition during ex vivo CD8+ T cell expansion facilitates the generation of polyfunctional T cells with stem cell memory-like traits. However, most therapeutic T cell products are generated from lymphocytes, containing CD4+ T cells that can affect CD8+ T cells dependent on the Th-subset. Here, we investigated the effect of AKT-inhibition on CD4+ T cells, during separate as well as total T cell expansions. Interestingly, ex vivo AKT-inhibition preserved the early memory phenotype of CD4+ T cells based on higher CD62L, CXCR4 and CCR7 expression. However, in the presence of AKT-inhibition, Th-differentiation was skewed toward more Th2-associated at the expense of Th1-associated cells. Importantly, the favorable effect of AKT-inhibition on the functionality of CD8+ T cells drastically diminished in the presence of CD4+ T cells. Moreover, also the expansion method influenced the effect of AKT-inhibition on CD8+ T cells. These findings indicate that the effect of AKT-inhibition on CD8+ T cells is dependent on cell composition and expansion strategy, where presence of CD4+ T cells as well as polyclonal stimulation impede the favorable effect of AKT-inhibition. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
16. Ex vivo AKT-inhibition facilitates generation of polyfunctional stem cell memory-like CD8+ T cells for adoptive immunotherapy
- Author
-
Mousset, Charlotte M., primary, Hobo, Willemijn, additional, Ji, Yun, additional, Fredrix, Hanny, additional, De Giorgi, Valeria, additional, Allison, Robert D., additional, Kester, Michel G. D., additional, Falkenburg, J. H. Frederik, additional, Schaap, Nicolaas P. M., additional, Jansen, Joop H., additional, Gattinoni, Luca, additional, Dolstra, Harry, additional, and van der Waart, Anniek B., additional
- Published
- 2018
- Full Text
- View/download PDF
17. Successful Transfer of Umbilical Cord Blood CD34+ Hematopoietic Stem and Progenitor-derived NK Cells in Older Acute Myeloid Leukemia Patients
- Author
-
Dolstra, Harry, primary, Roeven, Mieke W.H., additional, Spanholtz, Jan, additional, Hangalapura, Basav N., additional, Tordoir, Marleen, additional, Maas, Frans, additional, Leenders, Marij, additional, Bohme, Fenna, additional, Kok, Nina, additional, Trilsbeek, Carel, additional, Paardekooper, Jos, additional, van der Waart, Anniek B., additional, Westerweel, Peter E., additional, Snijders, Tjeerd J.F., additional, Cornelissen, Jan, additional, Bos, Gerard, additional, Pruijt, Hans F.M., additional, de Graaf, Aniek O., additional, van der Reijden, Bert A., additional, Jansen, Joop H., additional, van der Meer, Arnold, additional, Huls, Gerwin, additional, Cany, Jeannette, additional, Preijers, Frank, additional, Blijlevens, Nicole M.A., additional, and Schaap, Nicolaas M., additional
- Published
- 2017
- Full Text
- View/download PDF
18. A Phase I Study of Allogeneic Natural Killer Cell Therapy Generated from Cord Blood Hematopoietic Stem and Progenitor Cells in Elderly Acute Myeloid Leukemia Patients
- Author
-
Dolstra, Harry, primary, Roeven, Mieke W.H., additional, Spanholtz, Jan, additional, Hangalapura, Basav, additional, Tordoir, Marleen, additional, Maas, Frans, additional, Leenders, Marij, additional, Bohme, Fenna, additional, Kok, Nina, additional, Trilsbeek, Carel, additional, Paardekooper, Jos, additional, Van der Waart, Anniek B., additional, Westerweel, Peter, additional, Snijders, Tjeerd J.F., additional, Bos, Gerard M.J., additional, Cornelissen, Jan J., additional, Pruijt, Hans, additional, Huls, Gerwin, additional, de Graaf, Aniek, additional, van der Reijden, Bert A., additional, Blijlevens, Nicole M.A., additional, Jansen, Joop H., additional, van der Meer, Arnold, additional, Cany, Jeannette, additional, Preijers, Frank, additional, and Schaap, Nicolaas P.M., additional
- Published
- 2015
- Full Text
- View/download PDF
19. 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
-
Cany, Jeannette, primary, van der Waart, Anniek B, additional, Spanholtz, Jan, additional, Tordoir, Marleen, additional, Jansen, Joop H, additional, van der Voort, Robbert, additional, Schaap, Nicolaas M, additional, and Dolstra, Harry, additional
- Published
- 2015
- Full Text
- View/download PDF
20. Time toAkt
- Author
-
van der Waart, Anniek B, primary, Hobo, Willemijn, additional, and Dolstra, Harry, additional
- Published
- 2015
- Full Text
- View/download PDF
21. Akt Signalling Inhibition Promotes The Ex Vivo generation Of Minor Histocompatibility Antigen-Specific CD8+ Memory Stem T Cells
- Author
-
van der Waart, Anniek B., primary, van der Weem, Noortje, additional, Gattinoni, Luca, additional, Schaap, Nicolaas PM, additional, Voort, Robbert van der, additional, Hobo, Willemijn, additional, and Dolstra, Harry, additional
- Published
- 2013
- Full Text
- View/download PDF
22. Influence Of DNA Methyltransferese Inhibitors On The Anti-Leukemic Effect Of Umbilical Cord Blood Derived NK Cells Against Acute Myeloid Leukemia
- Author
-
Dolstra, Harry, primary, Cany, Jeannette, additional, van der Waart, Anniek B., additional, Tordoir, Marleen, additional, Hangalapura, Basav Nagaraj, additional, Spanholtz, Jan, additional, van der Voort, Robbert, additional, and Schaap, Nicolaas PM, additional
- Published
- 2013
- Full Text
- View/download PDF
23. Natural Killer Cells Generated from Cord Blood Hematopoietic Progenitor Cells Efficiently Target Bone Marrow-Residing Human Leukemia Cells in NOD/SCID/IL2Rgnull Mice
- Author
-
Cany, Jeannette, primary, van der Waart, Anniek B., additional, Tordoir, Marleen, additional, Franssen, Gerben M., additional, Hangalapura, Basav N., additional, de Vries, Jolanda, additional, Boerman, Otto, additional, Schaap, Nicolaas, additional, van der Voort, Robbert, additional, Spanholtz, Jan, additional, and Dolstra, Harry, additional
- Published
- 2013
- Full Text
- View/download PDF
24. Decreased Levels of Circulating IL17-Producing CD161+CCR6+ T Cells Are Associated with Graft-versus-Host Disease after Allogeneic Stem Cell Transplantation
- Author
-
van der Waart, Anniek B., primary, van der Velden, Walter J. F. M., additional, van Halteren, Astrid G. S., additional, Leenders, Marij J. L. G., additional, Feuth, Ton, additional, Blijlevens, Nicole M. A., additional, van der Voort, Robbert, additional, and Dolstra, Harry, additional
- Published
- 2012
- Full Text
- View/download PDF
25. Natural Killer Cells Generated from Cord Blood Hematopoietic Progenitor Cells Efficiently Target Bone Marrow-Residing Human Leukemia Cells in NOD/SCID/IL2Rgnull Mice.
- Author
-
Cany, Jeannette, van der Waart, Anniek B., Tordoir, Marleen, Franssen, Gerben M., Hangalapura, Basav N., de Vries, Jolanda, Boerman, Otto, Schaap, Nicolaas, van der Voort, Robbert, Spanholtz, Jan, and Dolstra, Harry
- Subjects
- *
KILLER cells , *PROGENITOR cells , *CORD blood , *HEMATOPOIETIC stem cells , *BONE marrow , *LEUKEMIA , *CANCER cells , *SEVERE combined immunodeficiency , *LABORATORY mice - Abstract
Natural killer (NK) cell-based adoptive immunotherapy is an attractive adjuvant treatment option for patients with acute myeloid leukemia. Recently, we reported a clinical-grade, cytokine-based culture method for the generation of NK cells from umbilical cord blood (UCB) CD34+ hematopoietic progenitor cells with high yield, purity and in vitro functionality. The present study was designed to evaluate the in vivo anti-leukemic potential of UCB-NK cells generated with our GMP-compliant culture system in terms of biodistribution, survival and cytolytic activity following adoptive transfer in immunodeficient NOD/SCID/IL2Rgnull mice. Using single photon emission computed tomography, we first demonstrated active migration of UCB-NK cells to bone marrow, spleen and liver within 24 h after infusion. Analysis of the chemokine receptor expression profile of UCB-NK cells matched in vivo findings. Particularly, a firm proportion of UCB-NK cells functionally expressed CXCR4, what could trigger BM homing in response to its ligand CXCL12. In addition, high expression of CXCR3 and CCR6 supported the capacity of UCB-NK cells to migrate to inflamed tissues via the CXCR3/CXCL10-11 and CCR6/CCL20 axis. Thereafter, we showed that low dose IL-15 mediates efficient survival, expansion and maturation of UCB-NK cells in vivo. Most importantly, we demonstrate that a single UCB-NK cell infusion combined with supportive IL-15 administration efficiently inhibited growth of human leukemia cells implanted in the femur of mice, resulting in significant prolongation of mice survival. These preclinical studies strongly support the therapeutic potential of ex vivo-generated UCB-NK cells in the treatment of myeloid leukemia after immunosuppressive chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
26. Decreased Levels of Circulating IL17-Producing CD161+CCR6+ T Cells Are Associated with Graft-versus-Host Disease after Allogeneic Stem Cell Transplantation.
- Author
-
van der Waart, Anniek B., van der Velden, Walter J. F. M., van Halteren, Astrid G. S., Leenders, Marij J. L. G., Feuth, Ton, Blijlevens, Nicole M. A., van der Voort, Robbert, and Dolstra, Harry
- Subjects
- *
T cells , *GRAFT versus host disease , *STEM cell transplantation , *CYCLOSPORINE , *IMMUNOSUPPRESSIVE agents , *TRANSPLANTATION of organs, tissues, etc. - Abstract
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, CD161hiCD8+ T cell numbers declined during tapering of Cyclosporine A (CsA), which can be explained by their initial growth advantage over CD161neg/lowCD8+ 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 CD161hiCD8+ T cells. In addition, these subsets from transplanted patients secreted high levels of IFNc 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+ coexpressing 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 [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
27. CD34+progenitor-derived NK cell and gemcitabine combination therapy increases killing of ovarian cancer cells in NOD/SCID/IL2Rgnullmice
- Author
-
Van der Meer, Jolien M.R., de Jonge, Paul K.J.D., van der Waart, Anniek B., Geerlings, Alexander C., Moonen, Jurgen P., Brummelman, Jolanda, de Klein, Janne, Vermeulen, Malou C., Maas, Ralph J.A., Schaap, Nicolaas P.M., Hoogstad-van Evert, Janneke S., Ottevanger, Petronella B., Jansen, Joop H., Hobo, Willemijn, and Dolstra, Harry
- Abstract
ABSTRACTCombining natural killer (NK) cell adoptive transfer with tumor-sensitizing chemotherapy is an attractive approach against recurrent ovarian cancer (OC), as OC is sensitive to NK cell-mediated immunity. Previously, we showed that CD34+hematopoietic progenitor cell (HPC)-derived NK cells can kill OC cells in vitroand inhibit OC tumor growth in mice. Here, we investigated the potential of HPC-NK cell therapy combined with chemotherapeutic gemcitabine (used in recurrent OC patients) against OC. We examined the phenotypical, functional, and cytotoxic effects of gemcitabine on HPC-NK cells and/or OC cells in vitroand in OC-bearing mice. To this end, we treated OC cells and/or HPC-NK cells with or without gemcitabine and analyzed the phenotype, cytokine production, and anti-tumor reactivity. We found that gemcitabine did not affect the phenotype and functionality of HPC-NK cells, while on OC cells expression of NK cell activating ligands and death receptors was upregulated. Although gemcitabine pre-treatment of OC cells did not improve the functionality of HPC-NK cells, importantly, HPC-NK cells and gemcitabine additively killed OC cells in vitro. Similarly, combined HPC-NK cell and gemcitabine treatment additively decreased tumor growth in OC-bearing mice. Collectively, our results indicate that combination therapy of HPC-NK cells and gemcitabine results in augmented OC killing in vitroand in vivo. This provides a rationale for exploring this therapeutic strategy in patients with recurrent OC.
- Published
- 2021
- Full Text
- View/download PDF
28. Ex vivo AKT-inhibition facilitates generation of polyfunctional stem cell memory-like CD8+ T cells for adoptive immunotherapy.
- Author
-
Mousset, Charlotte M., Hobo, Willemijn, Fredrix, Hanny, Jansen, Joop H., Dolstra, Harry, van der Waart, Anniek B., Ji, Yun, Gattinoni, Luca, De Giorgi, Valeria, Allison, Robert D., Kester, Michel G. D., Falkenburg, J. H. Frederik, and Schaap, Nicolaas P. M.
- Subjects
T cells ,CANCER ,PATIENTS ,ANTIGENS ,ADENOSINE triphosphatase ,PHENOTYPES ,TRANSCRIPTOMES - Abstract
Adoptive T cell therapy has shown clinical potential for patients with cancer, though effective treatment is dependent on longevity and potency of the exploited tumor-reactive T cells. Previously, we showed that ex vivo inhibition of AKT using the research compound Akt-inhibitor VIII retained differentiation and improved functionality of minor histocompatibility antigen (MiHA)-specific CD8
+ T cells. Here, we compared a panel of clinically applicable AKT-inhibitors with an allosteric or adenosine triphosphate-competitive mode of action. We analyzed phenotype, functionality, metabolism and transcriptome of AKT-inhibited CD8+ T cells using different T cell activation models. Most inhibitors facilitated T cell expansion while preserving an early memory phenotype, reflected by maintenance of CD62L, CCR7 and CXCR4 expression. Moreover, transcriptome profiling revealed that AKT-inhibited CD8+ T cells clustered closely to naturally occurring stem cell-memory CD8+ T cells, while control T cells resembled effector-memory T cells. Interestingly, AKT-inhibited CD8+ T cells showed enrichment of hypoxia-associated genes, which was consistent with enhanced glycolytic function. Notably, AKT-inhibition during MiHA-specific CD8+ T cell priming uncoupled preservation of early memory differentiation from ex vivo expansion. Furthermore, AKT-inhibited MiHA-specific CD8+ T cells showed increased polyfunctionality with co-secretion of IFN-γ and IL-2 upon antigen recall. Together, these data demonstrate that AKT-inhibitors with different modality of action promote the ex vivo generation of stem cell memory-like CD8+ T cells with a unique metabolic profile and retained polyfunctionality. Akt-inhibitor VIII and GDC-0068 outperformed other inhibitors, and are therefore promising candidates for ex vivo generation of superior tumor-reactive T cells for adoptive immunotherapy in cancer patients. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
29. Ex vivoAKT-inhibition facilitates generation of polyfunctional stem cell memory-like CD8+T cells for adoptive immunotherapy
- Author
-
Mousset, Charlotte M., Hobo, Willemijn, Ji, Yun, Fredrix, Hanny, De Giorgi, Valeria, Allison, Robert D., Kester, Michel G. D., Falkenburg, J. H. Frederik, Schaap, Nicolaas P. M., Jansen, Joop H., Gattinoni, Luca, Dolstra, Harry, and van der Waart, Anniek B.
- Abstract
ABSTRACTAdoptive T cell therapy has shown clinical potential for patients with cancer, though effective treatment is dependent on longevity and potency of the exploited tumor-reactive T cells. Previously, we showed that ex vivoinhibition of AKT using the research compound Akt-inhibitor VIII retained differentiation and improved functionality of minor histocompatibility antigen (MiHA)-specific CD8+T cells. Here, we compared a panel of clinically applicable AKT-inhibitors with an allosteric or adenosine triphosphate-competitive mode of action. We analyzed phenotype, functionality, metabolism and transcriptome of AKT-inhibited CD8+T cells using different T cell activation models. Most inhibitors facilitated T cell expansion while preserving an early memory phenotype, reflected by maintenance of CD62L, CCR7 and CXCR4 expression. Moreover, transcriptome profiling revealed that AKT-inhibited CD8+T cells clustered closely to naturally occurring stem cell-memory CD8+T cells, while control T cells resembled effector-memory T cells. Interestingly, AKT-inhibited CD8+T cells showed enrichment of hypoxia-associated genes, which was consistent with enhanced glycolytic function. Notably, AKT-inhibition during MiHA-specific CD8+T cell priming uncoupled preservation of early memory differentiation from ex vivoexpansion. Furthermore, AKT-inhibited MiHA-specific CD8+T cells showed increased polyfunctionality with co-secretion of IFN-γ and IL-2 upon antigen recall. Together, these data demonstrate that AKT-inhibitors with different modality of action promote the ex vivogeneration of stem cell memory-like CD8+T cells with a unique metabolic profile and retained polyfunctionality. Akt-inhibitor VIII and GDC-0068 outperformed other inhibitors, and are therefore promising candidates for ex vivogeneration of superior tumor-reactive T cells for adoptive immunotherapy in cancer patients.
- Published
- 2018
- Full Text
- View/download PDF
30. Time to Akt.
- Author
-
van der Waart, Anniek B, Hobo, Willemijn, and Dolstra, Harry
- Subjects
- *
T cells , *CANCER prevention , *CELLULAR therapy , *TUMORS , *CANCER relapse - Abstract
T cells are crucial players in the protection against cancer, and can be used in adoptive cell therapy to prevent or treat relapse. However, their state of differentiation determines their effectiveness, with early memory cells being the most favorable. Here, we discuss restraining of differentiation to engineer the ultimate tumor-reactive T cell. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
31. Akt Signalling Inhibition Promotes The Ex Vivogeneration Of Minor Histocompatibility Antigen-Specific CD8+ Memory Stem T Cells
- Author
-
van der Waart, Anniek B., van der Weem, Noortje, Gattinoni, Luca, Schaap, Nicolaas PM, Voort, Robbert van der, Hobo, Willemijn, and Dolstra, Harry
- 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 vitroinduction 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 vivofollowing T cell transfer. In this regard, the less-differentiated memory subset called stem cell memory T cells (TSCM) with superior in vivoexpansion, 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 vivoavailability and ex vivogeneration 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 TSCMphenotype defined by the expression of CD45RO, CCR7, CD27 and CD95. Though TSCMcells could be clearly detected within CMV-specific CD8+ T cells in allo-SCT patients, similar to healthy controls, no MiHA-specific TSCMcells 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 TCMand TSCM-like cells compared to almost entirely TEMcells 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 TEFFcells were highly functional, showing capacity to degranulate and produce IFNγ upon peptide restimulation. In conclusion, by inhibiting the Akt-pathway, in vitroCD8+ 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.
- Published
- 2013
- Full Text
- View/download PDF
32. Increased peritoneal TGF-β1 is associated with ascites-induced NK-cell dysfunction and reduced survival in high-grade epithelial ovarian cancer.
- Author
-
Maas RJA, Hoogstad-van Evert JS, Hagemans IM, Brummelman J, van Ens D, de Jonge PKJD, Hooijmaijers L, Mahajan S, van der Waart AB, Hermans CKJC, de Klein J, Woestenenk R, van Herwaarden AE, Schaap NPM, Rezaeifard S, Tauriello DVF, Zusterzeel PLM, Ottevanger N, Jansen JH, Hobo W, and Dolstra H
- Subjects
- Humans, Female, Middle Aged, Neoplasm Grading, Aged, Pyrazoles therapeutic use, Pyrazoles pharmacology, Quinolines, Transforming Growth Factor beta1 metabolism, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Carcinoma, Ovarian Epithelial immunology, Carcinoma, Ovarian Epithelial mortality, Ascites immunology, Ovarian Neoplasms immunology, Ovarian Neoplasms mortality, Ovarian Neoplasms pathology
- Abstract
Natural killer (NK) cell therapy represents an attractive immunotherapy approach against recurrent epithelial ovarian cancer (EOC), as EOC is sensitive to NK cell-mediated cytotoxicity. However, NK cell antitumor activity is dampened by suppressive factors in EOC patient ascites. Here, we integrated functional assays, soluble factor analysis, high-dimensional flow cytometry cellular component data and clinical parameters of advanced EOC patients to study the mechanisms of ascites-induced inhibition of NK cells. Using a suppression assay, we found that ascites from EOC patients strongly inhibits peripheral blood-derived NK cells and CD34+ progenitor-derived NK cells, albeit the latter were more resistant. Interestingly, we found that higher ascites-induced NK cell inhibition correlated with reduced progression-free and overall survival in EOC patients. Furthermore, we identified transforming growth factor (TGF)-β1 to correlate with ascites-induced NK cell dysfunction and reduced patient survival. In functional assays, we showed that proliferation and anti-tumor reactivity of CD34+ progenitor-derived NK cells are significantly affected by TGF-β1 exposure. Moreover, inhibition of TGF-β1 signaling with galunisertib partly restored NK cell functionality in some donors. For the cellular components, we showed that the secretome is associated with a different composition of CD45+ cells between ascites of EOC and benign reference samples with higher proportions of macrophages in the EOC patient samples. Furthermore, we revealed that higher TGF-β1 levels are associated with the presence of M2-like macrophages, B cell populations and T-regulatory cells in EOC patient ascites. These findings reveal that targeting TGF-β1 signaling could increase NK cell immune responses in high-grade EOC patients., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Maas, Hoogstad-van Evert, Hagemans, Brummelman, van Ens, de Jonge, Hooijmaijers, Mahajan, van der Waart, Hermans, de Klein, Woestenenk, van Herwaarden, Schaap, Rezaeifard, Tauriello, Zusterzeel, Ottevanger, Jansen, Hobo and Dolstra.)
- Published
- 2024
- Full Text
- View/download PDF
33. CD34 + progenitor-derived NK cell and gemcitabine combination therapy increases killing of ovarian cancer cells in NOD/SCID/IL2Rg null mice.
- Author
-
Van der Meer JMR, de Jonge PKJD, van der Waart AB, Geerlings AC, Moonen JP, Brummelman J, de Klein J, Vermeulen MC, Maas RJA, Schaap NPM, Hoogstad-van Evert JS, Ottevanger PB, Jansen JH, Hobo W, and Dolstra H
- Subjects
- Animals, Deoxycytidine analogs & derivatives, Female, Humans, Interleukin Receptor Common gamma Subunit, Killer Cells, Natural, Mice, Mice, Inbred NOD, Mice, SCID, Gemcitabine, Neoplasm Recurrence, Local, Ovarian Neoplasms drug therapy
- Abstract
Combining natural killer (NK) cell adoptive transfer with tumor-sensitizing chemotherapy is an attractive approach against recurrent ovarian cancer (OC), as OC is sensitive to NK cell-mediated immunity. Previously, we showed that CD34
+ hematopoietic progenitor cell (HPC)-derived NK cells can kill OC cells in vitro and inhibit OC tumor growth in mice. Here, we investigated the potential of HPC-NK cell therapy combined with chemotherapeutic gemcitabine (used in recurrent OC patients) against OC. We examined the phenotypical, functional, and cytotoxic effects of gemcitabine on HPC-NK cells and/or OC cells in vitro and in OC-bearing mice. To this end, we treated OC cells and/or HPC-NK cells with or without gemcitabine and analyzed the phenotype, cytokine production, and anti-tumor reactivity. We found that gemcitabine did not affect the phenotype and functionality of HPC-NK cells, while on OC cells expression of NK cell activating ligands and death receptors was upregulated. Although gemcitabine pre-treatment of OC cells did not improve the functionality of HPC-NK cells, importantly, HPC-NK cells and gemcitabine additively killed OC cells in vitro . Similarly, combined HPC-NK cell and gemcitabine treatment additively decreased tumor growth in OC-bearing mice. Collectively, our results indicate that combination therapy of HPC-NK cells and gemcitabine results in augmented OC killing in vitro and in vivo . This provides a rationale for exploring this therapeutic strategy in patients with recurrent OC., Competing Interests: None of the authors have any competing interests., (© 2021 The Author(s). Published with license by Taylor & Francis Group, LLC.)- Published
- 2021
- Full Text
- View/download PDF
34. Ex vivo AKT-inhibition facilitates generation of polyfunctional stem cell memory-like CD8 + T cells for adoptive immunotherapy.
- Author
-
Mousset CM, Hobo W, Ji Y, Fredrix H, De Giorgi V, Allison RD, Kester MGD, Falkenburg JHF, Schaap NPM, Jansen JH, Gattinoni L, Dolstra H, and van der Waart AB
- Abstract
Adoptive T cell therapy has shown clinical potential for patients with cancer, though effective treatment is dependent on longevity and potency of the exploited tumor-reactive T cells. Previously, we showed that ex vivo inhibition of AKT using the research compound Akt-inhibitor VIII retained differentiation and improved functionality of minor histocompatibility antigen (MiHA)-specific CD8
+ T cells. Here, we compared a panel of clinically applicable AKT-inhibitors with an allosteric or adenosine triphosphate-competitive mode of action. We analyzed phenotype, functionality, metabolism and transcriptome of AKT-inhibited CD8+ T cells using different T cell activation models. Most inhibitors facilitated T cell expansion while preserving an early memory phenotype, reflected by maintenance of CD62L, CCR7 and CXCR4 expression. Moreover, transcriptome profiling revealed that AKT-inhibited CD8+ T cells clustered closely to naturally occurring stem cell-memory CD8+ T cells, while control T cells resembled effector-memory T cells. Interestingly, AKT-inhibited CD8+ T cells showed enrichment of hypoxia-associated genes, which was consistent with enhanced glycolytic function. Notably, AKT-inhibition during MiHA-specific CD8+ T cell priming uncoupled preservation of early memory differentiation from ex vivo expansion. Furthermore, AKT-inhibited MiHA-specific CD8+ T cells showed increased polyfunctionality with co-secretion of IFN-γ and IL-2 upon antigen recall. Together, these data demonstrate that AKT-inhibitors with different modality of action promote the ex vivo generation of stem cell memory-like CD8+ T cells with a unique metabolic profile and retained polyfunctionality. Akt-inhibitor VIII and GDC-0068 outperformed other inhibitors, and are therefore promising candidates for ex vivo generation of superior tumor-reactive T cells for adoptive immunotherapy in cancer patients.- Published
- 2018
- Full Text
- View/download PDF
35. Successful Transfer of Umbilical Cord Blood CD34 + Hematopoietic Stem and Progenitor-derived NK Cells in Older Acute Myeloid Leukemia Patients.
- Author
-
Dolstra H, Roeven MWH, Spanholtz J, Hangalapura BN, Tordoir M, Maas F, Leenders M, Bohme F, Kok N, Trilsbeek C, Paardekooper J, van der Waart AB, Westerweel PE, Snijders TJF, Cornelissen J, Bos G, Pruijt HFM, de Graaf AO, van der Reijden BA, Jansen JH, van der Meer A, Huls G, Cany J, Preijers F, Blijlevens NMA, and Schaap NM
- Subjects
- Aged, Antigens, CD34 genetics, Antigens, CD34 immunology, Cord Blood Stem Cell Transplantation adverse effects, Female, Hematopoietic Stem Cell Transplantation adverse effects, Hematopoietic Stem Cells immunology, Hematopoietic Stem Cells metabolism, Humans, Interleukin-15 blood, Killer Cells, Natural transplantation, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute pathology, Male, Neoplasm Regression, Spontaneous pathology, Prognosis, Cell- and Tissue-Based Therapy, Cord Blood Stem Cell Transplantation methods, Hematopoietic Stem Cell Transplantation methods, Leukemia, Myeloid, Acute therapy
- Abstract
Purpose: Older acute myeloid leukemia (AML) patients have a poor prognosis; therefore, novel therapies are needed. Allogeneic natural killer (NK) cells have been adoptively transferred with promising clinical results. Here, we report the first-in-human study exploiting a unique scalable NK-cell product generated ex vivo from CD34
+ hematopoietic stem and progenitor cells (HSPC) from partially HLA-matched umbilical cord blood units. Experimental Design: Ten older AML patients in morphologic complete remission received an escalating HSPC-NK cell dose (between 3 and 30 × 106 /kg body weight) after lymphodepleting chemotherapy without cytokine boosting. Results: HSPC-NK cell products contained a median of 75% highly activated NK cells, with <1 × 104 T cells/kg and <3 × 105 B cells/kg body weight. HSPC-NK cells were well tolerated, and neither graft-versus-host disease nor toxicity was observed. Despite no cytokine boosting being given, transient HSPC-NK cell persistence was clearly found in peripheral blood up to 21% until day 8, which was accompanied by augmented IL15 plasma levels. Moreover, donor chimerism up to 3.5% was found in bone marrow. Interestingly, in vivo HSPC-NK cell maturation was observed, indicated by the rapid acquisition of CD16 and KIR expression, while expression of most activating receptors was sustained. Notably, 2 of 4 patients with minimal residual disease (MRD) in bone marrow before infusion became MRD negative (<0.1%), which lasted for 6 months. Conclusions: These findings indicate that HSPC-NK cell adoptive transfer is a promising, potential "off-the-shelf" translational immunotherapy approach in AML. Clin Cancer Res; 23(15); 4107-18. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
36. 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
-
Cany J, van der Waart AB, Spanholtz J, Tordoir M, Jansen JH, van der Voort R, Schaap NM, and Dolstra H
- 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 cell products demonstrated superior maturation potential, resulting in >70% positivity for CD16 and/or KIR within 2 weeks after infusion into NSG mice. We predict that higher functionality and faster in vivo maturation will favor HPC-NK cell alloreactivity toward malignant cells in patients, making this cytokine combination an attractive strategy to generate clinical HPC-NK cell products for cancer adoptive immunotherapy.- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.