191 results on '"P. Martens"'
Search Results
2. Gemtuzumab Ozogamicin Plus Intensive Chemotherapy for Patients with NPM1-Mutated Acute Myeloid Leukemia
- Author
-
Döhner, Hartmut, Weber, Daniela, Krzykalla, Julia, Fiedler, Walter, Kühn, Michael W.M., Schroeder, Thomas, Mayer, Karin, Lübbert, Michael, Wattad, Mohammad, Götze, Katharina, Fransecky, Lars, Koller, Elisabeth, Wulf, Gerald, Schleicher, Jan, Ringhoffer, Mark, Greil, Richard, Hertenstein, Bernd, Krauter, Jürgen, Martens, Uwe M., Nachbaur, David, Samra, Maisun Abu, Machherndl-Spandl, Sigrid, Basara, Nadezda, Leis, Claudia, Schrade, Anika, Kapp-Schwoerer, Silke, Bullinger, Lars, Thol, Felicitas R., Heuser, Michael, Paschka, Peter, Gaidzik, Verena I., Saadati, Maral, Benner, Axel, Schlenk, Richard F., Döhner, Konstanze, and Ganser, Arnold
- Published
- 2022
- Full Text
- View/download PDF
3. Dietary methionine starvation impairs acute myeloid leukemia progression
- Author
-
Cunningham, Alan, Erdem, Ayşegül, Alshamleh, Islam, Geugien, Marjan, Pruis, Maurien, Pereira-Martins, Diego Antonio, van den Heuvel, Fiona A. J., Wierenga, Albertus T. J., ten Berge, Hilde, Dennebos, Robin, van den Boom, Vincent, Hogeling, Shanna M., Weinhäuser, Isabel, Knops, Ruth, de Blaauw, Pim, Heiner-Fokkema, M. Rebecca, Woolthuis, Carolien, Günther, Ulrich L., Rego, Eduardo M., Martens, Joost H. A., Jansen, Joop H., Schwalbe, Harald, Huls, Gerwin, and Schuringa, Jan Jacob
- Abstract
Targeting altered tumor cell metabolism might provide an attractive opportunity for patients with acute myeloid leukemia (AML). An amino acid dropout screen on primary leukemic stem cells and progenitor populations revealed a number of amino acid dependencies, of which methionine was one of the strongest. By using various metabolite rescue experiments, nuclear magnetic resonance−based metabolite quantifications and 13C-tracing, polysomal profiling, and chromatin immunoprecipitation sequencing, we identified that methionine is used predominantly for protein translation and to provide methyl groups to histones via S-adenosylmethionine for epigenetic marking. H3K36me3 was consistently the most heavily impacted mark following loss of methionine. Methionine depletion also reduced total RNA levels, enhanced apoptosis, and induced a cell cycle block. Reactive oxygen species levels were not increased following methionine depletion, and replacement of methionine with glutathione or N-acetylcysteine could not rescue phenotypes, excluding a role for methionine in controlling redox balance control in AML. Although considered to be an essential amino acid, methionine can be recycled from homocysteine. We uncovered that this is primarily performed by the enzyme methionine synthase and only when methionine availability becomes limiting. In vivo, dietary methionine starvation was not only tolerated by mice, but also significantly delayed both cell line and patient-derived AML progression. Finally, we show that inhibition of the H3K36-specific methyltransferase SETD2 phenocopies much of the cytotoxic effects of methionine depletion, providing a more targeted therapeutic approach. In conclusion, we show that methionine depletion is a vulnerability in AML that can be exploited therapeutically, and we provide mechanistic insight into how cells metabolize and recycle methionine.
- Published
- 2022
- Full Text
- View/download PDF
4. Dietary methionine starvation impairs acute myeloid leukemia progression
- Author
-
Cunningham, Alan, Erdem, Ayşegül, Alshamleh, Islam, Geugien, Marjan, Pruis, Maurien, Pereira-Martins, Diego Antonio, van den Heuvel, Fiona A. J., Wierenga, Albertus T. J., ten Berge, Hilde, Dennebos, Robin, van den Boom, Vincent, Hogeling, Shanna M., Weinhäuser, Isabel, Knops, Ruth, de Blaauw, Pim, Heiner-Fokkema, M. Rebecca, Woolthuis, Carolien, Günther, Ulrich L., Rego, Eduardo M., Martens, Joost H. A., Jansen, Joop H., Schwalbe, Harald, Huls, Gerwin, and Schuringa, Jan Jacob
- Abstract
•Amino acid dropout screen reveals that AML cells highly depend on methionine, cysteine, arginine, glutamine, and lysine.•Dietary methionine removal impacts on the proteome, metabolome and epigenome, perturbing AML progression in vivo.
- Published
- 2022
- Full Text
- View/download PDF
5. Specific proteome changes in platelets from individuals with GATA1-, GFI1B-, and RUNX1-linked bleeding disorders
- Author
-
Van Bergen, M. G. J. M., Marneth, A. E., Hoogendijk, A. J., Van Alphen, F. P. J., Van den Akker, E., Laros-Van Gorkom, B. A. P., Hoeks, M., Simons, A., De Munnik, S. A., Janssen, J. J. W. M., Martens, J. H. A., Jansen, J. H., Meijer, A. B., and Van der Reijden, B. A.
- Abstract
Mutations in the transcription factors GATA binding factor 1 (GATA1), growth factor independence 1B (GFI1B), and Runt-related transcription factor 1 (RUNX1) cause familial platelet and bleeding disorders. Mutant platelets exhibit common abnormalities including an α-granule reduction resulting in a grayish appearance in blood smears. This suggests that similar pathways are deregulated by different transcription factor mutations. To identify common factors, full platelet proteomes from 11 individuals with mutant GATA1R216Q, GFI1BQ287*, RUNX1Q154Rfs, or RUNX1TD2-6 and 28 healthy controls were examined by label-free quantitative mass spectrometry. In total, 2875 platelet proteins were reliably quantified. Clustering analysis of more than 300 differentially expressed proteins revealed profound differences between cases and controls. Among cases, 44 of 143 significantly downregulated proteins were assigned to platelet function, hemostasis, and granule biology, in line with platelet dysfunction and bleedings. Remarkably, none of these proteins were significantly diminished in all affected cases. Similarly, no proteins were commonly overrepresented in all affected cases compared with controls. These data indicate that the studied transcription factor mutations alter platelet proteomes in distinct largely nonoverlapping manners. This work provides the quantitative landscape of proteins that affect platelet function when deregulated by mutated transcription factors in inherited bleeding disorders.
- Published
- 2021
- Full Text
- View/download PDF
6. Randomized multicenter trial of sirolimus vs prednisone as initial therapy for standard-risk acute GVHD: the BMT CTN 1501 trial
- Author
-
Pidala, Joseph, Hamadani, Mehdi, Dawson, Peter, Martens, Michael, Alousi, Amin M., Jagasia, Madan, Efebera, Yvonne A., Chhabra, Saurabh, Pusic, Iskra, Holtan, Shernan G., Ferrara, James L.M., Levine, John E., Mielcarek, Marco, Anasetti, Claudio, Antin, Joseph H., Bolaños-Meade, Javier, Howard, Alan, Logan, Brent R., Leifer, Eric S., Pritchard, Theresa S., Horowitz, Mary M., and MacMillan, Margaret L.
- Abstract
Clinical- and biomarker-based tools may identify a lower-risk acute graft-versus-host disease (GVHD) population amenable to novel, reduced-intensity treatments. Previous data suggest sirolimus may rival standard of care prednisone. We conducted a National Heart, Lung, and Blood Institute/National Cancer Institute-funded Blood and Marrow Transplant Clinical Trials Network multicenter, open-label, randomized phase 2 trial to estimate the difference in day 28 complete response (CR)/partial response (PR) rates for sirolimus vs prednisone as initial treatment of patients with standard risk (SR) acute GVHD as defined by the Minnesota (MN) GVHD Risk Score and Ann Arbor (AA1/2) biomarker status. A total of 127 MN-SR patients were randomized (1:1), and 122 were AA1/2 (sirolimus, n = 58; prednisone, n = 64). Others were AA3 (n = 4), or AA status missing (n = 1). The day 28 CR/PR rates were similar for sirolimus 64.8% (90% confidence interval [CI], 54.1%-75.5%) vs 73% (90% CI, 63.8%-82.2%) for prednisone. The day 28 rate of CR/PR with prednisone ≤0.25 mg/kg/day was significantly higher for sirolimus than prednisone (66.7% vs 31.7%; P< .001). No differences were detected in steroid-refractory acute GVHD, disease-free survival, relapse, nonrelapse mortality, or overall survival. Sirolimus was associated with reduced steroid exposure and hyperglycemia, reduced grade 2 to 3 infections, improvement in immune suppression discontinuation and patient-reported quality of life, and increased risk for thrombotic microangiopathy. For patients with clinical- and biomarker-based SR acute GVHD, sirolimus demonstrates similar overall initial treatment efficacy as prednisone. In addition, sirolimus therapy spares steroid exposure and allied toxicity, does not compromise long-term survival outcomes, and is associated with improved patient-reported quality of life. This trial was registered at www.clinicaltrials.govas #NCT02806947.
- Published
- 2020
- Full Text
- View/download PDF
7. Randomized multicenter trial of sirolimus vs prednisone as initial therapy for standard-risk acute GVHD: the BMT CTN 1501 trial
- Author
-
Pidala, Joseph, Hamadani, Mehdi, Dawson, Peter, Martens, Michael, Alousi, Amin M., Jagasia, Madan, Efebera, Yvonne A., Chhabra, Saurabh, Pusic, Iskra, Holtan, Shernan G., Ferrara, James L. M., Levine, John E., Mielcarek, Marco, Anasetti, Claudio, Antin, Joseph H., Bolaños-Meade, Javier, Howard, Alan, Logan, Brent R., Leifer, Eric S., Pritchard, Theresa S., Horowitz, Mary M., and MacMillan, Margaret L.
- Abstract
Clinical- and biomarker-based tools may identify a lower-risk acute graft-versus-host disease (GVHD) population amenable to novel, reduced-intensity treatments. Previous data suggest sirolimus may rival standard of care prednisone. We conducted a National Heart, Lung, and Blood Institute/National Cancer Institute-funded Blood and Marrow Transplant Clinical Trials Network multicenter, open-label, randomized phase 2 trial to estimate the difference in day 28 complete response (CR)/partial response (PR) rates for sirolimus vs prednisone as initial treatment of patients with standard risk (SR) acute GVHD as defined by the Minnesota (MN) GVHD Risk Score and Ann Arbor (AA1/2) biomarker status. A total of 127 MN-SR patients were randomized (1:1), and 122 were AA1/2 (sirolimus, n = 58; prednisone, n = 64). Others were AA3 (n = 4), or AA status missing (n = 1). The day 28 CR/PR rates were similar for sirolimus 64.8% (90% confidence interval [CI], 54.1%-75.5%) vs 73% (90% CI, 63.8%-82.2%) for prednisone. The day 28 rate of CR/PR with prednisone ≤0.25 mg/kg/day was significantly higher for sirolimus than prednisone (66.7% vs 31.7%; P < .001). No differences were detected in steroid-refractory acute GVHD, disease-free survival, relapse, nonrelapse mortality, or overall survival. Sirolimus was associated with reduced steroid exposure and hyperglycemia, reduced grade 2 to 3 infections, improvement in immune suppression discontinuation and patient-reported quality of life, and increased risk for thrombotic microangiopathy. For patients with clinical- and biomarker-based SR acute GVHD, sirolimus demonstrates similar overall initial treatment efficacy as prednisone. In addition, sirolimus therapy spares steroid exposure and allied toxicity, does not compromise long-term survival outcomes, and is associated with improved patient-reported quality of life. This trial was registered at www.clinicaltrials.gov as #NCT02806947.
- Published
- 2020
- Full Text
- View/download PDF
8. PHF19 promotes multiple myeloma tumorigenicity through PRC2 activation and broad H3K27me3 domain formation
- Author
-
Ren, Zhihong, Ahn, Jeong Hyun, Liu, Hequn, Tsai, Yi-Hsuan, Bhanu, Natarajan V., Koss, Brian, Allison, David F., Ma, Anqi, Storey, Aaron J., Wang, Ping, Mackintosh, Samuel G., Edmondson, Ricky D., Groen, Richard W. J., Martens, Anton C., Garcia, Benjamin A., Tackett, Alan J., Jin, Jian, Cai, Ling, Zheng, Deyou, and Wang, Gang Greg
- Abstract
Dysregulation of polycomb repressive complex 2 (PRC2) promotes oncogenesis partly through its enzymatic function for inducing trimethylation of histone H3 lysine 27 (H3K27me3). However, it remains to be determined how PRC2 activity is regulated in normal and diseased settings. We here report a PRC2-associated cofactor, PHD finger protein 19 (PHF19; also known as polycomb-like 3), as a crucial mediator of tumorigenicity in multiple myeloma (MM). Overexpression and/or genomic amplification of PHF19 is found associated with malignant progression of MM and plasma cell leukemia, correlating to worse treatment outcomes. Using various MM models, we demonstrated a critical requirement of PHF19 for tumor growth in vitro and in vivo. Mechanistically, PHF19-mediated oncogenic effect relies on its PRC2-interacting and chromatin-binding functions. Chromatin immunoprecipitation followed by sequencing profiling showed a critical role for PHF19 in maintaining the H3K27me3 landscape. PHF19 depletion led to loss of broad H3K27me3 domains, possibly due to impaired H3K27me3 spreading from cytosine guanine dinucleotide islands, which is reminiscent to the reported effect of an “onco”-histone mutation, H3K27 to methionine (H3K27M). RNA-sequencing–based transcriptome profiling in MM lines also demonstrated a requirement of PHF19 for optimal silencing of PRC2 targets, which include cell cycle inhibitors and interferon-JAK-STAT signaling genes critically involved in tumor suppression. Correlation studies using patient sample data sets further support a clinical relevance of the PHF19-regulated pathways. Lastly, we show that MM cells are generally sensitive to PRC2 inhibitors. Collectively, this study demonstrates that PHF19 promotes MM tumorigenesis through enhancing H3K27me3 deposition and PRC2’s gene-regulatory functions, lending support for PRC2 blockade as a means for MM therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
9. PHF19 promotes multiple myeloma tumorigenicity through PRC2 activation and broad H3K27me3 domain formation
- Author
-
Ren, Zhihong, Ahn, Jeong Hyun, Liu, Hequn, Tsai, Yi-Hsuan, Bhanu, Natarajan V., Koss, Brian, Allison, David F., Ma, Anqi, Storey, Aaron J., Wang, Ping, Mackintosh, Samuel G., Edmondson, Ricky D., Groen, RichardW.J., Martens, Anton C., Garcia, Benjamin A., Tackett, Alan J., Jin, Jian, Cai, Ling, Zheng, Deyou, and Wang, Gang Greg
- Abstract
Dysregulation of polycomb repressive complex 2 (PRC2) promotes oncogenesis partly through its enzymatic function for inducing trimethylation of histone H3 lysine 27 (H3K27me3). However, it remains to be determined how PRC2 activity is regulated in normal and diseased settings. We here report a PRC2-associated cofactor, PHD finger protein 19 (PHF19; also known as polycomb-like 3), as a crucial mediator of tumorigenicity in multiple myeloma (MM). Overexpression and/or genomic amplification of PHF19 is found associated with malignant progression of MM and plasma cell leukemia, correlating to worse treatment outcomes. Using various MM models, we demonstrated a critical requirement of PHF19 for tumor growth in vitro and in vivo. Mechanistically, PHF19-mediated oncogenic effect relies on its PRC2-interacting and chromatin-binding functions. Chromatin immunoprecipitation followed by sequencing profiling showed a critical role for PHF19 in maintaining the H3K27me3 landscape. PHF19 depletion led to loss of broad H3K27me3 domains, possibly due to impaired H3K27me3 spreading from cytosine guanine dinucleotide islands, which is reminiscent to the reported effect of an “onco”-histone mutation, H3K27 to methionine (H3K27M). RNA-sequencing–based transcriptome profiling in MM lines also demonstrated a requirement of PHF19 for optimal silencing of PRC2 targets, which include cell cycle inhibitors and interferon-JAK-STAT signaling genes critically involved in tumor suppression. Correlation studies using patient sample data sets further support a clinical relevance of the PHF19-regulated pathways. Lastly, we show that MM cells are generally sensitive to PRC2 inhibitors. Collectively, this study demonstrates that PHF19 promotes MM tumorigenesis through enhancing H3K27me3 deposition and PRC2's gene-regulatory functions, lending support for PRC2 blockade as a means for MM therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
10. Signaling mechanisms inducing hyporesponsiveness of phagocytes during systemic inflammation
- Author
-
Freise, Nicole, Burghard, Alina, Ortkras, Theresa, Daber, Niklas, Imam Chasan, Achmet, Jauch, Saskia-L., Fehler, Olesja, Hillebrand, Julia, Schakaki, Mosab, Rojas, Jessica, Grimbacher, Bodo, Vogl, Thomas, Hoffmeier, Andreas, Martens, Sven, Roth, Johannes, and Austermann, Judith
- Abstract
The inflammatory responsiveness of phagocytes to exogenous and endogenous stimuli is tightly regulated. This regulation plays an important role in systemic inflammatory response syndromes (SIRSs). In SIRSs, phagocytes initially develop a hyperinflammatory response, followed by a secondary state of hyporesponsiveness, a so-called “tolerance.” This hyporesponsiveness can be induced by endotoxin stimulation of Toll-like receptor 4 (TLR4), resulting in an ameliorated response after subsequent restimulation. This modification of inflammatory response patterns has been described as innate immune memory. Interestingly, tolerance can also be triggered by endogenous TLR4 ligands, such as the alarmins myeloid-related protein 8 (MRP8, S100A8) and MRP14 (S100A9), under sterile conditions. However, signaling pathways that trigger hyporesponsiveness of phagocytes in clinically relevant diseases are only barely understood. Through our work, we have now identified 2 main signaling cascades that are activated during MRP-induced tolerance of phagocytes. We demonstrate that the phosphatidylinositol 3-kinase/AKT/GSK-3ß pathway interferes with NF-?B–driven gene expression and that inhibition of GSK-3ß mimics tolerance in vivo. Moreover, we identified interleukin-10–triggered activation of transcription factors STAT3 and BCL-3 as master regulators of MRP-induced tolerance. Accordingly, patients with dominant-negative STAT3 mutations show no tolerance development. In a clinically relevant condition of systemic sterile stress, cardiopulmonary bypass surgery, we confirmed the initial induction of MRP expression and the tolerance induction of monocytes associated with nuclear translocation of STAT3 and BCL-3 as relevant mechanisms. Our data indicate that the use of pharmacological JAK-STAT inhibitors may be promising targets for future therapeutic approaches to prevent complications associated with secondary hyporesponsiveness during SIRS.
- Published
- 2019
- Full Text
- View/download PDF
11. Signaling mechanisms inducing hyporesponsiveness of phagocytes during systemic inflammation
- Author
-
Freise, Nicole, Burghard, Alina, Ortkras, Theresa, Daber, Niklas, Imam Chasan, Achmet, Jauch, Saskia-L., Fehler, Olesja, Hillebrand, Julia, Schakaki, Mosab, Rojas, Jessica, Grimbacher, Bodo, Vogl, Thomas, Hoffmeier, Andreas, Martens, Sven, Roth, Johannes, and Austermann, Judith
- Abstract
The inflammatory responsiveness of phagocytes to exogenous and endogenous stimuli is tightly regulated. This regulation plays an important role in systemic inflammatory response syndromes (SIRSs). In SIRSs, phagocytes initially develop a hyperinflammatory response, followed by a secondary state of hyporesponsiveness, a so-called “tolerance.” This hyporesponsiveness can be induced by endotoxin stimulation of Toll-like receptor 4 (TLR4), resulting in an ameliorated response after subsequent restimulation. This modification of inflammatory response patterns has been described as innate immune memory. Interestingly, tolerance can also be triggered by endogenous TLR4 ligands, such as the alarmins myeloid-related protein 8 (MRP8, S100A8) and MRP14 (S100A9), under sterile conditions. However, signaling pathways that trigger hyporesponsiveness of phagocytes in clinically relevant diseases are only barely understood. Through our work, we have now identified 2 main signaling cascades that are activated during MRP-induced tolerance of phagocytes. We demonstrate that the phosphatidylinositol 3-kinase/AKT/GSK-3β pathway interferes with NF-κB–driven gene expression and that inhibition of GSK-3β mimics tolerance in vivo. Moreover, we identified interleukin-10–triggered activation of transcription factors STAT3 and BCL-3 as master regulators of MRP-induced tolerance. Accordingly, patients with dominant-negative STAT3 mutations show no tolerance development. In a clinically relevant condition of systemic sterile stress, cardiopulmonary bypass surgery, we confirmed the initial induction of MRP expression and the tolerance induction of monocytes associated with nuclear translocation of STAT3 and BCL-3 as relevant mechanisms. Our data indicate that the use of pharmacological JAK-STAT inhibitors may be promising targets for future therapeutic approaches to prevent complications associated with secondary hyporesponsiveness during SIRS.
- Published
- 2019
- Full Text
- View/download PDF
12. Chronic lymphocytic leukemia cells impair mitochondrial fitness in CD8+ T cells and impede CAR T-cell efficacy
- Author
-
van Bruggen, Jaco A. C., Martens, Anne W. J., Fraietta, Joseph A., Hofland, Tom, Tonino, Sanne H., Eldering, Eric, Levin, Mark-David, Siska, Peter J., Endstra, Sanne, Rathmell, Jeffrey C., June, Carl H., Porter, David L., Melenhorst, J. Joseph, Kater, Arnon P., and van der Windt, Gerritje J. W.
- Abstract
In chronic lymphocytic leukemia (CLL), acquired T-cell dysfunction impedes development of effective immunotherapeutic strategies, through as-yet unresolved mechanisms. We have previously shown that CD8+ T cells in CLL exhibit impaired activation and reduced glucose uptake after stimulation. CD8+ T cells in CLL patients are chronically exposed to leukemic B cells, which potentially impacts metabolic homeostasis resulting in aberrant metabolic reprogramming upon stimulation. Here, we report that resting CD8+ T cells in CLL have reduced intracellular glucose transporter 1 (GLUT1) reserves, and have an altered mitochondrial metabolic profile as displayed by increased mitochondrial respiration, membrane potential, and levels of reactive oxygen species. This coincided with decreased levels of peroxisome proliferator-activated receptor ? coactivator 1-a, and in line with that, CLL-derived CD8+ T cells showed impaired mitochondrial biogenesis upon stimulation. In search of a therapeutic correlate of these findings, we analyzed mitochondrial biogenesis in CD19-directed chimeric antigen receptor (CAR) CD8+ T cells prior to infusion in CLL patients (who were enrolled in NCT01747486 and NCT01029366 [https://clinicaltrials.gov]). Interestingly, in cases with a subsequent complete response, the infused CD8+ CAR T cells had increased mitochondrial mass compared with nonresponders, which positively correlated with the expansion and persistence of CAR T cells. Our findings demonstrate that GLUT1 reserves and mitochondrial fitness of CD8+ T cells are impaired in CLL. Therefore, boosting mitochondrial biogenesis in CAR T cells might improve the efficacy of CAR T-cell therapy and other emerging cellular immunotherapies.
- Published
- 2019
- Full Text
- View/download PDF
13. Chronic lymphocytic leukemia cells impair mitochondrial fitness in CD8+T cells and impede CAR T-cell efficacy
- Author
-
van Bruggen, Jaco A.C., Martens, Anne W.J., Fraietta, Joseph A., Hofland, Tom, Tonino, Sanne H., Eldering, Eric, Levin, Mark-David, Siska, Peter J., Endstra, Sanne, Rathmell, Jeffrey C., June, Carl H., Porter, David L., Melenhorst, J. Joseph, Kater, Arnon P., and van der Windt, Gerritje J.W.
- Abstract
In chronic lymphocytic leukemia (CLL), acquired T-cell dysfunction impedes development of effective immunotherapeutic strategies, through as-yet unresolved mechanisms. We have previously shown that CD8+T cells in CLL exhibit impaired activation and reduced glucose uptake after stimulation. CD8+T cells in CLL patients are chronically exposed to leukemic B cells, which potentially impacts metabolic homeostasis resulting in aberrant metabolic reprogramming upon stimulation. Here, we report that resting CD8+T cells in CLL have reduced intracellular glucose transporter 1 (GLUT1) reserves, and have an altered mitochondrial metabolic profile as displayed by increased mitochondrial respiration, membrane potential, and levels of reactive oxygen species. This coincided with decreased levels of peroxisome proliferator-activated receptor γ coactivator 1-α, and in line with that, CLL-derived CD8+T cells showed impaired mitochondrial biogenesis upon stimulation. In search of a therapeutic correlate of these findings, we analyzed mitochondrial biogenesis in CD19-directed chimeric antigen receptor (CAR) CD8+T cells prior to infusion in CLL patients (who were enrolled in NCT01747486 and NCT01029366 [https://clinicaltrials.gov]). Interestingly, in cases with a subsequent complete response, the infused CD8+CAR T cells had increased mitochondrial mass compared with nonresponders, which positively correlated with the expansion and persistence of CAR T cells. Our findings demonstrate that GLUT1 reserves and mitochondrial fitness of CD8+T cells are impaired in CLL. Therefore, boosting mitochondrial biogenesis in CAR T cells might improve the efficacy of CAR T-cell therapy and other emerging cellular immunotherapies.
- Published
- 2019
- Full Text
- View/download PDF
14. Midostaurin added to chemotherapy and continued single-agent maintenance therapy in acute myeloid leukemia with FLT3-ITD
- Author
-
Schlenk, Richard F., Weber, Daniela, Fiedler, Walter, Salih, Helmut R., Wulf, Gerald, Salwender, Hans, Schroeder, Thomas, Kindler, Thomas, Lübbert, Michael, Wolf, Dominik, Westermann, Jörg, Kraemer, Doris, Götze, Katharina S., Horst, Heinz-August, Krauter, Jürgen, Girschikofsky, Michael, Ringhoffer, Mark, Südhoff, Thomas, Held, Gerhard, Derigs, Hans-Günter, Schroers, Roland, Greil, Richard, Grießhammer, Martin, Lange, Elisabeth, Burchardt, Alexander, Martens, Uwe, Hertenstein, Bernd, Marretta, Lore, Heuser, Michael, Thol, Felicitas, Gaidzik, Verena I., Herr, Wolfgang, Krzykalla, Julia, Benner, Axel, Döhner, Konstanze, Ganser, Arnold, Paschka, Peter, and Döhner, Hartmut
- Abstract
Patients with acute myeloid leukemia (AML) and a FLT3internal tandem duplication (ITD) have poor outcomes to current treatment. A phase 2 hypothesis-generating trial was conducted to determine whether the addition of the multitargeted kinase inhibitor midostaurin to intensive chemotherapy followed by allogeneic hematopoietic cell transplantation (alloHCT) and single-agent maintenance therapy of 12 months is feasible and favorably influences event-free survival (EFS) compared with historical controls. Patients 18 to 70 years of age with newly diagnosed AML and centrally confirmed FLT3-ITD were eligible: 284 patients were treated, including 198 younger (18-60 years) and 86 older (61-70 years) patients. Complete remission (CR) rate, including CR with incomplete hematological recovery (CRi) after induction therapy, was 76.4% (younger, 75.8%; older, 77.9%). The majority of patients in CR/CRi proceeded to alloHCT (72.4%). Maintenance therapy was started in 97 patients (34%): 75 after alloHCT and 22 after consolidation with high-dose cytarabine (HiDAC). Median time receiving maintenance therapy was 9 months after alloHCT and 10.5 months after HiDAC; premature termination was mainly a result of nonrelapse causes (gastrointestinal toxicity and infections). EFS and overall survival at 2 years were 39% (95% confidence interval [CI], 33%-47%) and 34% (95% CI, 24%-47%) and 53% (95% CI, 46%-61%) and 46% (95% CI, 35%-59%) in younger and older patients, respectively. EFS was evaluated in comparison with 415 historical controls treated within 5 prospective trials. Propensity score-weighted analysis revealed a significant improvement of EFS by midostaurin (hazard ratio [HR], 0.58; 95% CI, 0.48-0.70; P< .001) overall and in older patients (HR, 0.42; 95% CI, 0.29-0.61). The study was registered at www.clinicaltrials.govas #NCT01477606.
- Published
- 2019
- Full Text
- View/download PDF
15. Midostaurin added to chemotherapy and continued single-agent maintenance therapy in acute myeloid leukemia with FLT3-ITD
- Author
-
Schlenk, Richard F., Weber, Daniela, Fiedler, Walter, Salih, Helmut R., Wulf, Gerald, Salwender, Hans, Schroeder, Thomas, Kindler, Thomas, Lübbert, Michael, Wolf, Dominik, Westermann, Jörg, Kraemer, Doris, Götze, Katharina S., Horst, Heinz-August, Krauter, Jürgen, Girschikofsky, Michael, Ringhoffer, Mark, Südhoff, Thomas, Held, Gerhard, Derigs, Hans-Günter, Schroers, Roland, Greil, Richard, Grießhammer, Martin, Lange, Elisabeth, Burchardt, Alexander, Martens, Uwe, Hertenstein, Bernd, Marretta, Lore, Heuser, Michael, Thol, Felicitas, Gaidzik, Verena I., Herr, Wolfgang, Krzykalla, Julia, Benner, Axel, Döhner, Konstanze, Ganser, Arnold, Paschka, Peter, and Döhner, Hartmut
- Abstract
Patients with acute myeloid leukemia (AML) and a FLT3 internal tandem duplication (ITD) have poor outcomes to current treatment. A phase 2 hypothesis-generating trial was conducted to determine whether the addition of the multitargeted kinase inhibitor midostaurin to intensive chemotherapy followed by allogeneic hematopoietic cell transplantation (alloHCT) and single-agent maintenance therapy of 12 months is feasible and favorably influences event-free survival (EFS) compared with historical controls. Patients 18 to 70 years of age with newly diagnosed AML and centrally confirmed FLT3-ITD were eligible: 284 patients were treated, including 198 younger (18-60 years) and 86 older (61-70 years) patients. Complete remission (CR) rate, including CR with incomplete hematological recovery (CRi) after induction therapy, was 76.4% (younger, 75.8%; older, 77.9%). The majority of patients in CR/CRi proceeded to alloHCT (72.4%). Maintenance therapy was started in 97 patients (34%): 75 after alloHCT and 22 after consolidation with high-dose cytarabine (HiDAC). Median time receiving maintenance therapy was 9 months after alloHCT and 10.5 months after HiDAC; premature termination was mainly a result of nonrelapse causes (gastrointestinal toxicity and infections). EFS and overall survival at 2 years were 39% (95% confidence interval [CI], 33%-47%) and 34% (95% CI, 24%-47%) and 53% (95% CI, 46%-61%) and 46% (95% CI, 35%-59%) in younger and older patients, respectively. EFS was evaluated in comparison with 415 historical controls treated within 5 prospective trials. Propensity score-weighted analysis revealed a significant improvement of EFS by midostaurin (hazard ratio [HR], 0.58; 95% CI, 0.48-0.70; P < .001) overall and in older patients (HR, 0.42; 95% CI, 0.29-0.61). The study was registered at www.clinicaltrials.gov as #NCT01477606.
- Published
- 2019
- Full Text
- View/download PDF
16. Immunological Subgroups Predicting Efficacy of Elotuzumab in Multiple Myeloma Patients: Insights from the GMMG-HD6 Clinical Trial
- Author
-
Ton, Gigi Nu Hoang Quy, Kriegsmann, Katharina, Awwad, Mohamed H.S., Benner, Axel, Bertsch, Uta, Besemer, Britta, Hänel, Mathias, Fenk, Roland, Schlenzka, Jana, Munder, Markus, Dürig, Jan, Blau, Igor Wolfgang, Huhn, Stefanie, Hose, Dirk, Jauch, Anna, Neubauer, Andreas, Weinhold, Niels, Scheid, Christof, Schroers, Roland, von Metzler, Ivana, Schieferdecker, Aneta, Thomalla, Jörg, Reimer, Peter, Mahlberg, Rolf, Graeven, Ullrich, Kremers, Stephan, Martens, Uwe, Kunz, Christian, Hensel, Manfred, Seidel-Glätzer, Andrea, Weisel, Katja, Salwender, Hans, Müller-Tidow, Carsten, Raab, Marc S., Goldschmidt, Hartmut, Mai, Elias K., and Hundemer, Michael
- Abstract
Introduction: Elotuzumab, a monoclonal antibody targeting SLAM family member 7 protein (SLAMF7) on multiple myeloma (MM) cells, has shown promise in relapsed/refractory MM when combined with immunomodulatory agents. However, its effectiveness in newly diagnosed patients did not show improved progression-free survival (PFS) nor overall survival. In a previous study we demonstrated that elotuzumab specifically depleted high SLAMF7 expressing regulatory CD8 +T cells by macrophage induced antibody dependent phagocytosis. This study aimed to identify immunological predictive factors for elotuzumab efficacy, enabling improved risk stratification and personalized treatment decisions based on the detection and quantification of SLAMF7-positive T cell subsets.
- Published
- 2023
- Full Text
- View/download PDF
17. Patient-Reported Outcomes of BMT CTN 1703: A Randomized Phase III Study for Gvhd Prophylaxis - a Quality of Life Evaluation
- Author
-
Holtan, Shernan G., Martens, Michael J, Al Malki, Monzr M., Efebera, Yvonne, Kitko, Carrie L., Reshef, Ran, Rezvani, Andrew R., Shaffer, Brian C., Solh, Melhem M., Yao, Janny M., Runaas, Lyndsey, Elmariah, Hany, Larkin, Karilyn T., El Jurdi, Najla H, Gooptu, Mahasweta, Loren, Alison W, Hall, Aric C., Alousi, Amin, Jamy, Omer, Clark, William, Kean, Leslie, Bhatt, Ami S., Perales, Miguel-Angel, Applegate, Kristy, WU, Juan, Liefer, Eric, DiFronzo, Nancy, Jones, Richard J., Horowitz, Mary M., Mattila, Deborah, Hamadani, Mehdi, and Bolanos-Meade, Javier
- Abstract
Introduction
- Published
- 2023
- Full Text
- View/download PDF
18. Machine Learning Validates Risk Biomarkers of Chronic Graft-Versus-Host Disease in 936 Patients from BMT CTN 0201 & 1202 Cohorts
- Author
-
Martens, Michael J, Kou, Jianqun, Logan, Brent R., and Paczesny, Sophie
- Abstract
Introduction
- Published
- 2023
- Full Text
- View/download PDF
19. Hematology Electronic Consultation As a Means for Effective Management of Iron Deficiency Anemia
- Author
-
King, Hannah, Benedetti, Genevieve, Shatzel, Joseph J., Deloughery, Thomas G, and Martens, Kylee L
- Abstract
Introduction: The use of non-visit electronic consultation (e-consult) can improve patient access to hematology care when an urgent in-person evaluation is not required. Prior studies have confirmed that e-consults have been rapidly adopted in certain settings, can resolve queries without the need for in-person follow-up, and, in turn, lead to an increase in total consult volume institutionally. Iron deficiency is a common reason for e-consult, however utilization trends, the efficacy and safety of iron repletion in this context, and the avoidance of face-to-face hematology consultation remain largely unknown.
- Published
- 2023
- Full Text
- View/download PDF
20. Somatic Mutations and DNA Hypermethylation at Enhancers and Promoters Identify Distinct Subtypes within Lower-Risk Myelodysplastic Syndromes
- Author
-
Rombaut, David, Tekath, Tobias, Sandmann, Sarah, Crouch, Simon, de Graaf, Aniek O., Kosmider, Olivier, Tobiasson, Magnus, Lennartsson, Andreas, Van der Reijden, Bert A., Park, Sophie, D'Aveni, Maud, Slama, Bohrane, Clappier, Emmanuelle, Fenaux, Pierre, Ades, Lionel, van de Loosdrecht, Arjan A., Langemeijer, Saskia MC, Symeonidis, Argiris S., Čermák, Jaroslav, Preudhomme, Claude, Savic, Aleksandar, Germing, Ulrich, Stauder, Reinhard, Bowen, David T., Van Marrewijk, Corine J., Bernard, Elsa, Smith, Alexandra, Painter, Daniel, de Witte, Theo J.M., Hellstrom Lindberg, Eva, Varghese, Julian, Dugas, Martin, Martens, Joost H.A., Malcovati, Luca, Jansen, Joop H., and Fontenay, Michaela
- Abstract
Myelodysplastic syndromes (MDS) are a group of heterogeneous disorders caused by the accumulation of somatic mutations in the hematopoietic stem and progenitor compartment. Besides del(5q), SF3B1or TP53mutations, referred to as defining genetic abnormalities, mutation patterning hardly structured the classification of MDS. Mutations in epigenetic factors occur early in the development of clonal hematopoiesis leading to precocious alterations of DNA methylation implicated in oncogenesis. Convergent DNA methylation patterns related to both mutations and microenvironment imprinting may induce specific gene expression profiles and contribute to phenotypic variations. To refine a pathophysiological classification of lower-risk MDS, we combined genetic profiling to DNA methylation and transcriptome data.
- Published
- 2023
- Full Text
- View/download PDF
21. Pseudoprogression As Adverse Event of Chimeric Antigen Receptor T Cell Therapy
- Author
-
Schroeder, Torsten, Martens, Tjark, Chitadze, Guranda, Yalcin, Fatih, Trautmann, Heiko, Brüggemann, Monika, Heinen, Christine, Nakov, Philipp, Albici, Anca Maria, Spath, Nicolas, Sprenger, Lukas, Kramp, Laura Jane, Ritgen, Matthias, Wellnitz, Dominique, Simon, Maciej, Lippross, Sebastian, Iaccarino, Ingram, Klapper, Wolfram, Fransecky, Lars, Valerius, Thomas, Schub, Natalie, Pott, Christiane, Baldus, Claudia D, and Stoelzel, Friedrich
- Abstract
CAR-T-cell therapy (CARTCT) expands the range of therapeutic options for patients with hematologic malignancies. While complications such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are well known, pseudoprogression (PP) is until now mainly described after checkpoint inhibition. Some reports on CARTCT induced PP (CARTiPP) exist distinguishing it from progressive disease, while only few reports focus on acute local complications. Considering the paucity of data about CARTiPP, we aimed to assess patients treated in our center. We defined CARTiPP as initial volume increase of malignant manifestations within ten days of CARTCT followed by tumor regression without another genesis being more probable. Distinction from true progression of malignancy can be achieved either retrospectively or by histopathological findings.
- Published
- 2023
- Full Text
- View/download PDF
22. Correction of Anemia with Intravenous Iron Mitigates Adverse Maternal Outcomes in Pregnancy
- Author
-
Benson, Ashley E, Martens, Kylee L, Ryan, Kimberly S, Lo, Jamie O, Garg, Bharti, Chobrutskiy, Boris I, Deloughery, Thomas G, and Shatzel, Joseph J.
- Abstract
Background and Aim:Iron deficiency anemia (IDA) is common in pregnancy and associated with both maternal and fetal morbidity. Despite its high prevalence and associated complications, there is a lack of consensus regarding the treatment of iron deficiency (ID) in pregnant individuals. In spite of the higher efficacy and tolerance of intravenous (IV) iron compared to oral formulations, obstetric guidelines reserve its use for IDA refractory to oral treatment, even in late gestation. This study sought to determine maternal and neonatal outcomes in individuals receiving IV iron supplementation antepartum, stratified by severity of IDA.
- Published
- 2023
- Full Text
- View/download PDF
23. A Multi-Center Biologic Assignment Trial Comparing Reduced Intensity Allogeneic Hematopoietic Cell Transplantation to Hypomethylating Therapy or Best Supportive Care in Patients Aged 50-75 with Advanced Myelodysplastic Syndrome: Blood and Marrow Transplant Clinical Trials Network Study 1102
- Author
-
Nakamura, Ryotaro, Saber, Wael, Martens, Michael J, Ramirez, Alyssa, Scott, Bart L., Oran, Betul, Leifer, Eric, Tamari, Roni, Mishra, Asmita, Maziarz, Richard T., McGuirk, Joseph P., Westervelt, Peter, Vasu, Sumithra, Patnaik, Mrinal M., Kamble, Rammurti, Forman, Stephen J., Sekeres, Mikkael A., Appelbaum, Frederick R., Mendizabal, Adam M., Logan, Brent, Horowitz, Mary M., and Cutler, Corey
- Published
- 2020
- Full Text
- View/download PDF
24. Quality of Life in Patients Undergoing Double Umbilical Cord Blood Vs. Haploidentical Marrow Transplantation: A QOL Analysis Report of BMT CTN 1101
- Author
-
El Jurdi, Najla, Martens, Michael, Brunstein, Claudio G., O'Donnell, Paul, Lee, Stephanie J., D'Souza, Anita, Logan, Brent, Hong, Sanghee, Sandhu, Karam, Shapiro, Roman M, Singh, Anurag K., Horowitz, Mary M., and Hamilton, Betty K.
- Published
- 2022
- Full Text
- View/download PDF
25. Performance of D-Dimers in Patients with Prior History of Venous Thromboembolism Based on Anticoagulation Status
- Author
-
Mai, Vicky, Martens, Emily S.L., Righini, Marc, Schulman, Sam, Thiruganasambandamoorthy, Venkatesh, Kahn, Susan, Bates, Veronica, Pecarskie, Amanda, Kovacs, Michael J., Visser, Shaun, Shivakumar, Sudeep P, Tan, Melanie, Rodger, Marc A., Scarvelis, Dimitrios, Delluc, Aurelien, Girard, Philippe, Huisman, Menno V, Wells, Philip S., Klok, Frederikus A, and Le Gal, Gregoire
- Published
- 2022
- Full Text
- View/download PDF
26. Clinical Outcomes of Patients Referred for Asymptomatic Neutropenia at a Single Academic Center: A Focus on Racial Disparities
- Author
-
Oyogoa, Emmanuella, Martens, Kylee L, McMurry, Hannah Stowe, Lavasseur, Corinne Marie, Olson, Sven R, Deloughery, Thomas G, and Shatzel, Joseph J
- Published
- 2022
- Full Text
- View/download PDF
27. Utilization of Natural Language Processing in Venous Thromboembolism Identification
- Author
-
Avery, Jonathan, Martens, Kylee L, Nguyen, Daniel, Basom, Ryan, Lee, Stephanie, Garcia, David A., Rojas Hernandez, Cristhiam Mauricio, and Li, Ang
- Published
- 2022
- Full Text
- View/download PDF
28. Utilization of Natural Language Processing in Venous Thromboembolism Identification
- Author
-
Avery, Jonathan, Martens, Kylee L, Nguyen, Daniel, Basom, Ryan, Lee, Stephanie, Garcia, David A., Rojas Hernandez, Cristhiam Mauricio, and Li, Ang
- Published
- 2022
- Full Text
- View/download PDF
29. Quality of Life in Patients Undergoing Double Umbilical Cord Blood Vs. Haploidentical Marrow Transplantation: A QOL Analysis Report of BMT CTN 1101
- Author
-
El Jurdi, Najla, Martens, Michael, Brunstein, Claudio G., O'Donnell, Paul, Lee, Stephanie J., D'Souza, Anita, Logan, Brent, Hong, Sanghee, Sandhu, Karam, Shapiro, Roman M, Singh, Anurag K., Horowitz, Mary M., and Hamilton, Betty K.
- Published
- 2022
- Full Text
- View/download PDF
30. Performance of D-Dimers in Patients with Prior History of Venous Thromboembolism Based on Anticoagulation Status
- Author
-
Mai, Vicky, Martens, Emily S.L., Righini, Marc, Schulman, Sam, Thiruganasambandamoorthy, Venkatesh, Kahn, Susan, Bates, Veronica, Pecarskie, Amanda, Kovacs, Michael J., Visser, Shaun, Shivakumar, Sudeep P, Tan, Melanie, Rodger, Marc A., Scarvelis, Dimitrios, Delluc, Aurelien, Girard, Philippe, Huisman, Menno V, Wells, Philip S., Klok, Frederikus A, and Le Gal, Gregoire
- Published
- 2022
- Full Text
- View/download PDF
31. Clinical Outcomes of Patients Referred for Asymptomatic Neutropenia at a Single Academic Center: A Focus on Racial Disparities
- Author
-
Oyogoa, Emmanuella, Martens, Kylee L, McMurry, Hannah Stowe, Lavasseur, Corinne Marie, Olson, Sven R, Deloughery, Thomas G, and Shatzel, Joseph J
- Published
- 2022
- Full Text
- View/download PDF
32. Increasing use of allogeneic hematopoietic cell transplantation in patients aged 70 years and older in the United States
- Author
-
Muffly, Lori, Pasquini, Marcelo C., Martens, Michael, Brazauskas, Ruta, Zhu, Xiaochun, Adekola, Kehinde, Aljurf, Mahmoud, Ballen, Karen K., Bajel, Ashish, Baron, Frederic, Battiwalla, Minoo, Beitinjaneh, Amer, Cahn, Jean-Yves, Carabasi, Mathew, Chen, Yi-Bin, Chhabra, Saurabh, Ciurea, Stefan, Copelan, Edward, D'Souza, Anita, Edwards, John, Foran, James, Freytes, Cesar O., Fung, Henry C., Gale, Robert Peter, Giralt, Sergio, Hashmi, Shahrukh K., Hildebrandt, Gerhard C., Ho, Vincent, Jakubowski, Ann, Lazarus, Hillard, Luskin, Marlise R., Martino, Rodrigo, Maziarz, Richard, McCarthy, Philip, Nishihori, Taiga, Olin, Rebecca, Olsson, Richard F., Pawarode, Attaphol, Peres, Edward, Rezvani, Andrew R., Rizzieri, David, Savani, Bipin N., Schouten, Harry C., Sabloff, Mitchell, Seftel, Matthew, Seo, Sachiko, Sorror, Mohamed L., Szer, Jeff, Wirk, Baldeep M., Wood, William A., and Artz, Andrew
- Abstract
In this study, we evaluated trends and outcomes of allogeneic hematopoietic cell transplantation (HCT) in adults ≥70 years with hematologic malignancies across the United States. Adults ≥70 years with a hematologic malignancy undergoing first allogeneic HCT in the United States between 2000 and 2013 and reported to the Center for International Blood and Marrow Transplant Research were eligible. Transplant utilization and transplant outcomes, including overall survival (OS), progression-free survival (PFS), and transplant-related mortality (TRM) were studied. One thousand one hundred and six patients ≥70 years underwent HCT across 103 transplant centers. The number and proportion of allografts performed in this population rose markedly over the past decade, accounting for 0.1% of transplants in 2000 to 3.85% (N = 298) in 2013. Acute myeloid leukemia and myelodysplastic syndromes represented the most common disease indications. Two-year OS and PFS significantly improved over time (OS: 26% [95% confidence interval (CI), 21% to 33%] in 2000-2007 to 39% [95% CI, 35% to 42%] in 2008-2013, P< .001; PFS: 22% [16% to 28%] in 2000-2007 to 32% [95% CI, 29% to 36%] in 2008-2013, P= .003). Two-year TRM ranged from 33% to 35% and was unchanged over time (P= .54). Multivariable analysis of OS in the modern era of 2008-2013 revealed higher comorbidity by HCT comorbidity index ≥3 (hazard ratio [HR], 1.27; P= .006), umbilical cord blood graft (HR, 1.97; P= .0002), and myeloablative conditioning (HR, 1.61; P= .0002) as adverse factors. Over the past decade, utilization and survival after allogeneic transplant have increased in patients ≥70 years. Select adults ≥70 years with hematologic malignancies should be considered for transplant.
- Published
- 2017
- Full Text
- View/download PDF
33. Increasing use of allogeneic hematopoietic cell transplantation in patients aged 70 years and older in the United States
- Author
-
Muffly, Lori, Pasquini, Marcelo C., Martens, Michael, Brazauskas, Ruta, Zhu, Xiaochun, Adekola, Kehinde, Aljurf, Mahmoud, Ballen, Karen K., Bajel, Ashish, Baron, Frederic, Battiwalla, Minoo, Beitinjaneh, Amer, Cahn, Jean-Yves, Carabasi, Mathew, Chen, Yi-Bin, Chhabra, Saurabh, Ciurea, Stefan, Copelan, Edward, D’Souza, Anita, Edwards, John, Foran, James, Freytes, Cesar O., Fung, Henry C., Gale, Robert Peter, Giralt, Sergio, Hashmi, Shahrukh K., Hildebrandt, Gerhard C., Ho, Vincent, Jakubowski, Ann, Lazarus, Hillard, Luskin, Marlise R., Martino, Rodrigo, Maziarz, Richard, McCarthy, Philip, Nishihori, Taiga, Olin, Rebecca, Olsson, Richard F., Pawarode, Attaphol, Peres, Edward, Rezvani, Andrew R., Rizzieri, David, Savani, Bipin N., Schouten, Harry C., Sabloff, Mitchell, Seftel, Matthew, Seo, Sachiko, Sorror, Mohamed L., Szer, Jeff, Wirk, Baldeep M., Wood, William A., and Artz, Andrew
- Abstract
In this study, we evaluated trends and outcomes of allogeneic hematopoietic cell transplantation (HCT) in adults ≥70 years with hematologic malignancies across the United States. Adults ≥70 years with a hematologic malignancy undergoing first allogeneic HCT in the United States between 2000 and 2013 and reported to the Center for International Blood and Marrow Transplant Research were eligible. Transplant utilization and transplant outcomes, including overall survival (OS), progression-free survival (PFS), and transplant-related mortality (TRM) were studied. One thousand one hundred and six patients ≥70 years underwent HCT across 103 transplant centers. The number and proportion of allografts performed in this population rose markedly over the past decade, accounting for 0.1% of transplants in 2000 to 3.85% (N = 298) in 2013. Acute myeloid leukemia and myelodysplastic syndromes represented the most common disease indications. Two-year OS and PFS significantly improved over time (OS: 26% [95% confidence interval (CI), 21% to 33%] in 2000-2007 to 39% [95% CI, 35% to 42%] in 2008-2013, P < .001; PFS: 22% [16% to 28%] in 2000-2007 to 32% [95% CI, 29% to 36%] in 2008-2013, P = .003). Two-year TRM ranged from 33% to 35% and was unchanged over time (P = .54). Multivariable analysis of OS in the modern era of 2008-2013 revealed higher comorbidity by HCT comorbidity index ≥3 (hazard ratio [HR], 1.27; P = .006), umbilical cord blood graft (HR, 1.97; P = .0002), and myeloablative conditioning (HR, 1.61; P = .0002) as adverse factors. Over the past decade, utilization and survival after allogeneic transplant have increased in patients ≥70 years. Select adults ≥70 years with hematologic malignancies should be considered for transplant.
- Published
- 2017
- Full Text
- View/download PDF
34. Establishing human leukemia xenograft mouse models by implanting human bone marrow–like scaffold-based niches
- Author
-
Antonelli, Antonella, Noort, Willy A., Jaques, Jenny, de Boer, Bauke, de Jong-Korlaar, Regina, Brouwers-Vos, Annet Z., Lubbers-Aalders, Linda, van Velzen, Jeroen F., Bloem, Andries C., Yuan, Huipin, de Bruijn, Joost D., Ossenkoppele, Gert J., Martens, Anton C. M., Vellenga, Edo, Groen, Richard W. J., and Schuringa, Jan Jacob
- Abstract
To begin to understand the mechanisms that regulate self-renewal, differentiation, and transformation of human hematopoietic stem cells or to evaluate the efficacy of novel treatment modalities, stem cells need to be studied in their own species-specific microenvironment. By implanting ceramic scaffolds coated with human mesenchymal stromal cells into immune-deficient mice, we were able to mimic the human bone marrow niche. Thus, we have established a human leukemia xenograft mouse model in which a large cohort of patient samples successfully engrafted, which covered all of the important genetic and risk subgroups. We found that by providing a humanized environment, stem cell self-renewal properties were better maintained as determined by serial transplantation assays and genome-wide transcriptome studies, and less clonal drift was observed as determined by exome sequencing. The human leukemia xenograft mouse models that we have established here will serve as an excellent resource for future studies aimed at exploring novel therapeutic approaches.
- Published
- 2016
- Full Text
- View/download PDF
35. Establishing human leukemia xenograft mouse models by implanting human bone marrow–like scaffold-based niches
- Author
-
Antonelli, Antonella, Noort, Willy A., Jaques, Jenny, de Boer, Bauke, de Jong-Korlaar, Regina, Brouwers-Vos, Annet Z., Lubbers-Aalders, Linda, van Velzen, Jeroen F., Bloem, Andries C., Yuan, Huipin, de Bruijn, Joost D., Ossenkoppele, Gert J., Martens, Anton C.M., Vellenga, Edo, Groen, Richard W.J., and Schuringa, Jan Jacob
- Abstract
To begin to understand the mechanisms that regulate self-renewal, differentiation, and transformation of human hematopoietic stem cells or to evaluate the efficacy of novel treatment modalities, stem cells need to be studied in their own species-specific microenvironment. By implanting ceramic scaffolds coated with human mesenchymal stromal cells into immune-deficient mice, we were able to mimic the human bone marrow niche. Thus, we have established a human leukemia xenograft mouse model in which a large cohort of patient samples successfully engrafted, which covered all of the important genetic and risk subgroups. We found that by providing a humanized environment, stem cell self-renewal properties were better maintained as determined by serial transplantation assays and genome-wide transcriptome studies, and less clonal drift was observed as determined by exome sequencing. The human leukemia xenograft mouse models that we have established here will serve as an excellent resource for future studies aimed at exploring novel therapeutic approaches.
- Published
- 2016
- Full Text
- View/download PDF
36. Post-Transplant Cyclophosphamide, Tacrolimus, and Mycophenolate Mofetil As the New Standard for Graft-Versus-Host Disease (GVHD) Prophylaxis in Reduced Intensity Conditioning: Results from Phase III BMT CTN 1703
- Author
-
Holtan, Shernan G, Hamadani, Mehdi, WU, Juan, AL Malki, Monzr M., Runaas, Lyndsey, Elmariah, Hany, Rezvani, Andrew R., Gooptu, Mahasweta, Larkin, Karilyn T., Shaffer, Brian C., El Jurdi, Najla H, Loren, Alison W., Solh, Melhem, Hall, Aric C., Alousi, Amin M, Jamy, Omer H, Perales, Miguel-Angel, Yao, Janny M., Applegate, Kristy, Bhatt, Ami S., Kean, Leslie S., Efebera, Yvonne A., Kitko, Carrie Lynn, Reshef, Ran, Clark, William, DiFronzo, Nancy L., Henderson, Lori, Jones, Richard J., Liefer, Eric, Martens, Michael J, Horowitz, Mary M., and Bolanos-Meade, Javier
- Published
- 2022
- Full Text
- View/download PDF
37. Post-Transplant Cyclophosphamide, Tacrolimus, and Mycophenolate Mofetil As the New Standard for Graft-Versus-Host Disease (GVHD) Prophylaxis in Reduced Intensity Conditioning: Results from Phase III BMT CTN 1703
- Author
-
Holtan, Shernan G, Hamadani, Mehdi, WU, Juan, AL Malki, Monzr M., Runaas, Lyndsey, Elmariah, Hany, Rezvani, Andrew R., Gooptu, Mahasweta, Larkin, Karilyn T., Shaffer, Brian C., El Jurdi, Najla H, Loren, Alison W., Solh, Melhem, Hall, Aric C., Alousi, Amin M, Jamy, Omer H, Perales, Miguel-Angel, Yao, Janny M., Applegate, Kristy, Bhatt, Ami S., Kean, Leslie S., Efebera, Yvonne A., Kitko, Carrie Lynn, Reshef, Ran, Clark, William, DiFronzo, Nancy L., Henderson, Lori, Jones, Richard J., Liefer, Eric, Martens, Michael J, Horowitz, Mary M., and Bolanos-Meade, Javier
- Published
- 2022
- Full Text
- View/download PDF
38. Enforced differentiation of Dnmt3a-null bone marrow leads to failure with c-Kit mutations driving leukemic transformation
- Author
-
Celik, Hamza, Mallaney, Cates, Kothari, Alok, Ostrander, Elizabeth L., Eultgen, Elizabeth, Martens, Andrew, Miller, Christopher A., Hundal, Jasreet, Klco, Jeffery M., and Challen, Grant A.
- Abstract
Genome sequencing studies of patient samples have implicated the involvement of various components of the epigenetic machinery in myeloid diseases, including the de novo DNA methyltransferase DNMT3A. We have recently shown that Dnmt3a is essential for hematopoietic stem cell differentiation. Here, we investigated the effect of loss of Dnmt3a on hematopoietic transformation by forcing the normally quiescent hematopoietic stem cells to divide in vivo. Mice transplanted with Dnmt3a-null bone marrow in the absence of wild-type support cells succumbed to bone marrow failure (median survival, 328 days) characteristic of myelodysplastic syndromes with symptoms including anemia, neutropenia, bone marrow hypercellularity, and splenomegaly with myeloid infiltration. Two out of 25 mice developed myeloid leukemia with >20% blasts in the blood and bone marrow. Four out of 25 primary mice succumbed to myeloproliferative disorders, some of which progressed to secondary leukemia after long latency. Exome sequencing identified cooperating c-Kit mutations found only in the leukemic samples. Ectopic introduction of c-Kit variants into a Dnmt3a-deficient background produced acute leukemia with a short latency (median survival, 67 days). Our data highlight crucial roles of Dnmt3a in normal and malignant hematopoiesis and suggest that a major role for this enzyme is to facilitate developmental progression of progenitor cells at multiple decision checkpoints.
- Published
- 2015
- Full Text
- View/download PDF
39. Enforced differentiation of Dnmt3a-null bone marrow leads to failure with c-Kit mutations driving leukemic transformation
- Author
-
Celik, Hamza, Mallaney, Cates, Kothari, Alok, Ostrander, Elizabeth L., Eultgen, Elizabeth, Martens, Andrew, Miller, Christopher A., Hundal, Jasreet, Klco, Jeffery M., and Challen, Grant A.
- Abstract
Genome sequencing studies of patient samples have implicated the involvement of various components of the epigenetic machinery in myeloid diseases, including the de novo DNA methyltransferase DNMT3A. We have recently shown that Dnmt3ais essential for hematopoietic stem cell differentiation. Here, we investigated the effect of loss of Dnmt3aon hematopoietic transformation by forcing the normally quiescent hematopoietic stem cells to divide in vivo. Mice transplanted with Dnmt3a-null bone marrow in the absence of wild-type support cells succumbed to bone marrow failure (median survival, 328 days) characteristic of myelodysplastic syndromes with symptoms including anemia, neutropenia, bone marrow hypercellularity, and splenomegaly with myeloid infiltration. Two out of 25 mice developed myeloid leukemia with >20% blasts in the blood and bone marrow. Four out of 25 primary mice succumbed to myeloproliferative disorders, some of which progressed to secondary leukemia after long latency. Exome sequencing identified cooperating c-Kitmutations found only in the leukemic samples. Ectopic introduction of c-Kit variants into a Dnmt3a-deficient background produced acute leukemia with a short latency (median survival, 67 days). Our data highlight crucial roles of Dnmt3ain normal and malignant hematopoiesis and suggest that a major role for this enzyme is to facilitate developmental progression of progenitor cells at multiple decision checkpoints.
- Published
- 2015
- Full Text
- View/download PDF
40. Time-resolved characterization of cAMP/PKA-dependent signaling reveals that platelet inhibition is a concerted process involving multiple signaling pathways
- Author
-
Beck, Florian, Geiger, Jörg, Gambaryan, Stepan, Veit, Johannes, Vaudel, Marc, Nollau, Peter, Kohlbacher, Oliver, Martens, Lennart, Walter, Ulrich, Sickmann, Albert, and Zahedi, René P.
- Abstract
One of the most important physiological platelet inhibitors is endothelium-derived prostacyclin which stimulates the platelet cyclic adenosine monophosphate/protein kinase A (cAMP/PKA)–signaling cascade and inhibits virtually all platelet-activating key mechanisms. Using quantitative mass spectrometry, we analyzed time-resolved phosphorylation patterns in human platelets after treatment with iloprost, a stable prostacyclin analog, for 0, 10, 30, and 60 seconds to characterize key mediators of platelet inhibition and activation in 3 independent biological replicates. We quantified over 2700 different phosphorylated peptides of which 360 were significantly regulated upon stimulation. This comprehensive and time-resolved analysis indicates that platelet inhibition is a multipronged process involving different kinases and phosphatases as well as many previously unanticipated proteins and pathways.
- Published
- 2014
- Full Text
- View/download PDF
41. γ9 and δ2CDR3 domains regulate functional avidity of T cells harboring γ9δ2TCRs
- Author
-
Gründer, Cordula, van Dorp, Suzanne, Hol, Samantha, Drent, Esther, Straetemans, Trudy, Heijhuurs, Sabine, Scholten, Kirsten, Scheper, Wouter, Sebestyen, Zsolt, Martens, Anton, Strong, Roland, and Kuball, Jürgen
- Abstract
Immunotherapy with innate immune cells has recently evoked broad interest as a novel treatment option for cancer patients. γ9δ2T cells in particular are emerging as an innate cell population with high frequency and strong antitumor reactivity, which makes them and their receptors promising candidates for immune interventions. However, clinical trials have so far reported only limited tumor control by adoptively transferred γ9δ2T cells. As a potential explanation for this lack of efficacy, we found unexpectedly high variability in tumor recognition within the physiologic human γ9δ2T-cell repertoire, which is substantially regulated by the CDR3 domains of individual γ9δ2TCRs. In the present study, we demonstrate that the reported molecular requirements of CDR3 domains to interact with target cells shape the physiologic γ9δ2T-cell repertoire and, most likely, limit the protective and therapeutic antitumor efficacy of γ9δ2T cells. Based on these findings, we propose combinatorial-γδTCR-chain exchange as an efficient method for designing high-affinity γ9δ2TCRs that mediate improved antitumor responses when expressed in αβT cells both in vitro and in vivo in a humanized mouse model.
- Published
- 2012
- Full Text
- View/download PDF
42. γ9 and δ2CDR3 domains regulate functional avidity of T cells harboring γ9δ2TCRs
- Author
-
Gründer, Cordula, van Dorp, Suzanne, Hol, Samantha, Drent, Esther, Straetemans, Trudy, Heijhuurs, Sabine, Scholten, Kirsten, Scheper, Wouter, Sebestyen, Zsolt, Martens, Anton, Strong, Roland, and Kuball, Jürgen
- Abstract
Immunotherapy with innate immune cells has recently evoked broad interest as a novel treatment option for cancer patients. γ9δ2T cells in particular are emerging as an innate cell population with high frequency and strong antitumor reactivity, which makes them and their receptors promising candidates for immune interventions. However, clinical trials have so far reported only limited tumor control by adoptively transferred γ9δ2T cells. As a potential explanation for this lack of efficacy, we found unexpectedly high variability in tumor recognition within the physiologic human γ9δ2T-cell repertoire, which is substantially regulated by the CDR3 domains of individual γ9δ2TCRs. In the present study, we demonstrate that the reported molecular requirements of CDR3 domains to interact with target cells shape the physiologic γ9δ2T-cell repertoire and, most likely, limit the protective and therapeutic antitumor efficacy of γ9δ2T cells. Based on these findings, we propose combinatorial-γδTCR-chain exchange as an efficient method for designing high-affinity γ9δ2TCRs that mediate improved antitumor responses when expressed in αβT cells both in vitro and in vivo in a humanized mouse model.
- Published
- 2012
- Full Text
- View/download PDF
43. ERG and FLI1 binding sites demarcate targets for aberrant epigenetic regulation by AML1-ETO in acute myeloid leukemia
- Author
-
Martens, Joost H. A., Mandoli, Amit, Simmer, Femke, Wierenga, Bart-Jan, Saeed, Sadia, Singh, Abhishek A., Altucci, Lucia, Vellenga, Edo, and Stunnenberg, Hendrik G.
- Abstract
ERG and FLI1 are closely related members of the ETS family of transcription factors and have been identified as essential factors for the function and maintenance of normal hematopoietic stem cells. Here genome-wide analysis revealed that both ERG and FLI1 occupy similar genomic regions as AML1-ETO in t(8;21) AMLs and identified ERG/FLI1 as proteins that facilitate binding of oncofusion protein complexes. In addition, we demonstrate that ERG and FLI1 bind the RUNX1 promoter and that shRNA-mediated silencing of ERG leads to reduced expression of RUNX1 and AML1-ETO, consistent with a role of ERG in transcriptional activation of these proteins. Finally, we identify H3 acetylation as the epigenetic mark preferentially associated with ETS factor binding. This intimate connection between ERG/FLI1 binding and H3 acetylation implies that one of the molecular strategies of oncofusion proteins, such as AML1-ETO and PML-RAR-α, involves the targeting of histone deacetylase activities to ERG/FLI1 bound hematopoietic regulatory sites. Together, these results highlight the dual importance of ETS factors in t(8;21) leukemogenesis, both as transcriptional regulators of the oncofusion protein itself as well as proteins that facilitate AML1-ETO binding.
- Published
- 2012
- Full Text
- View/download PDF
44. ERG and FLI1 binding sites demarcate targets for aberrant epigenetic regulation by AML1-ETO in acute myeloid leukemia
- Author
-
Martens, Joost H.A., Mandoli, Amit, Simmer, Femke, Wierenga, Bart-Jan, Saeed, Sadia, Singh, Abhishek A., Altucci, Lucia, Vellenga, Edo, and Stunnenberg, Hendrik G.
- Abstract
ERG and FLI1 are closely related members of the ETS family of transcription factors and have been identified as essential factors for the function and maintenance of normal hematopoietic stem cells. Here genome-wide analysis revealed that both ERG and FLI1 occupy similar genomic regions as AML1-ETO in t(8;21) AMLs and identified ERG/FLI1 as proteins that facilitate binding of oncofusion protein complexes. In addition, we demonstrate that ERG and FLI1 bind the RUNX1 promoter and that shRNA-mediated silencing of ERG leads to reduced expression of RUNX1 and AML1-ETO, consistent with a role of ERG in transcriptional activation of these proteins. Finally, we identify H3 acetylation as the epigenetic mark preferentially associated with ETS factor binding. This intimate connection between ERG/FLI1 binding and H3 acetylation implies that one of the molecular strategies of oncofusion proteins, such as AML1-ETO and PML-RAR-α, involves the targeting of histone deacetylase activities to ERG/FLI1 bound hematopoietic regulatory sites. Together, these results highlight the dual importance of ETS factors in t(8;21) leukemogenesis, both as transcriptional regulators of the oncofusion protein itself as well as proteins that facilitate AML1-ETO binding.
- Published
- 2012
- Full Text
- View/download PDF
45. The first comprehensive and quantitative analysis of human platelet protein composition allows the comparative analysis of structural and functional pathways
- Author
-
Burkhart, Julia M., Vaudel, Marc, Gambaryan, Stepan, Radau, Sonja, Walter, Ulrich, Martens, Lennart, Geiger, Jörg, Sickmann, Albert, and Zahedi, René P.
- Abstract
Antiplatelet treatment is of fundamental importance in combatting functions/dysfunction of platelets in the pathogenesis of cardiovascular and inflammatory diseases. Dysfunction of anucleate platelets is likely to be completely attributable to alterations in posttranslational modifications and protein expression. We therefore examined the proteome of platelets highly purified from fresh blood donations, using elaborate protocols to ensure negligible contamination by leukocytes, erythrocytes, and plasma. Using quantitative mass spectrometry, we created the first comprehensive and quantitative human platelet proteome, comprising almost 4000 unique proteins, estimated copy numbers for ∼ 3700 of those, and assessed intersubject (4 donors) as well as intrasubject (3 different blood samples from 1 donor) variations of the proteome. For the first time, our data allow for a systematic and weighted appraisal of protein networks and pathways in human platelets, and indicate the feasibility of differential and comprehensive proteome analyses from small blood donations. Because 85% of the platelet proteome shows no variation between healthy donors, this study represents the starting point for disease-oriented platelet proteomics. In the near future, comprehensive and quantitative comparisons between normal and well-defined dysfunctional platelets, or between platelets obtained from donors at various stages of chronic cardiovascular and inflammatory diseases will be feasible.
- Published
- 2012
- Full Text
- View/download PDF
46. Chromatin accessibility, p300, and histone acetylation define PML-RARα and AML1-ETO binding sites in acute myeloid leukemia
- Author
-
Saeed, Sadia, Logie, Colin, Francoijs, Kees-Jan, Frigè, Gianmaria, Romanenghi, Mauro, Nielsen, Fiona G., Raats, Lianne, Shahhoseini, Maryam, Huynen, Martijn, Altucci, Lucia, Minucci, Saverio, Martens, Joost H. A., and Stunnenberg, Hendrik G.
- Abstract
Chromatin accessibility plays a key role in regulating cell type specific gene expression during hematopoiesis but has also been suggested to be aberrantly regulated during leukemogenesis. To understand the leukemogenic chromatin signature, we analyzed acute promyelocytic leukemia, a subtype of leukemia characterized by the expression of RARα-fusion proteins, such as PML-RARα. We used nuclease accessibility sequencing in cell lines as well as patient blasts to identify accessible DNA elements and identified > 100 000 accessible regions in each case. Using ChIP-seq, we identified H2A.Z as a histone modification generally associated with these accessible regions, whereas unsupervised clustering analysis of other chromatin features, including DNA methylation, H2A.Zac, H3ac, H3K9me3, H3K27me3, and the regulatory factor p300, distinguished 6 distinct clusters of accessible sites, each with a characteristic functional makeup. Of these, PML-RARα binding was found specifically at accessible chromatin regions characterized by p300 binding and hypoacetylated histones. Identifying regions with a similar epigenetic make up in t(8;21) acute myeloid leukemia (AML) cells, another subtype of AMLs, revealed that these regions are occupied by the oncofusion protein AML1-ETO. Together, our results suggest that oncofusion proteins localize to accessible regions and that chromatin accessibility together with p300 binding and histone acetylation characterize AML1-ETO and PML-RARα binding sites.
- Published
- 2012
- Full Text
- View/download PDF
47. Reconstructing the human hematopoietic niche in immunodeficient mice: opportunities for studying primary multiple myeloma
- Author
-
Groen, Richard W.J., Noort, Willy A., Raymakers, Reinier A., Prins, Henk-Jan, Aalders, Linda, Hofhuis, Frans M., Moerer, Petra, van Velzen, Jeroen F., Bloem, Andries C., van Kessel, Berris, Rozemuller, Henk, van Binsbergen, Ellen, Buijs, Arjan, Yuan, Huipin, de Bruijn, Joost D., de Weers, Michel, Parren, Paul W.H.I., Schuringa, Jan Jacob, Lokhorst, Henk M., Mutis, Tuna, and Martens, Anton C.M.
- Abstract
Interactions within the hematopoietic niche in the BM microenvironment are essential for maintenance of the stem cell pool. In addition, this niche is thought to serve as a sanctuary site for malignant progenitors during chemotherapy. Therapy resistance induced by interactions with the BM microenvironment is a major drawback in the treatment of hematologic malignancies and bone-metastasizing solid tumors. To date, studying these interactions was hampered by the lack of adequate in vivo models that simulate the human situation. In the present study, we describe a unique human-mouse hybrid model that allows engraftment and outgrowth of normal and malignant hematopoietic progenitors by implementing a technology for generating a human bone environment. Using luciferase gene marking of patient-derived multiple myeloma cells and bioluminescent imaging, we were able to follow pMM cells outgrowth and to visualize the effect of treatment. Therapeutic interventions in this model resulted in equivalent drug responses as observed in the corresponding patients. This novel human-mouse hybrid model creates unprecedented opportunities to investigate species-specific microenvironmental influences on normal and malignant hematopoietic development, and to develop and personalize cancer treatment strategies.
- Published
- 2012
- Full Text
- View/download PDF
48. Reconstructing the human hematopoietic niche in immunodeficient mice: opportunities for studying primary multiple myeloma
- Author
-
Groen, Richard W. J., Noort, Willy A., Raymakers, Reinier A., Prins, Henk-Jan, Aalders, Linda, Hofhuis, Frans M., Moerer, Petra, van Velzen, Jeroen F., Bloem, Andries C., van Kessel, Berris, Rozemuller, Henk, van Binsbergen, Ellen, Buijs, Arjan, Yuan, Huipin, de Bruijn, Joost D., de Weers, Michel, Parren, Paul W. H. I., Schuringa, Jan Jacob, Lokhorst, Henk M., Mutis, Tuna, and Martens, Anton C. M.
- Abstract
Interactions within the hematopoietic niche in the BM microenvironment are essential for maintenance of the stem cell pool. In addition, this niche is thought to serve as a sanctuary site for malignant progenitors during chemotherapy. Therapy resistance induced by interactions with the BM microenvironment is a major drawback in the treatment of hematologic malignancies and bone-metastasizing solid tumors. To date, studying these interactions was hampered by the lack of adequate in vivo models that simulate the human situation. In the present study, we describe a unique human-mouse hybrid model that allows engraftment and outgrowth of normal and malignant hematopoietic progenitors by implementing a technology for generating a human bone environment. Using luciferase gene marking of patient-derived multiple myeloma cells and bioluminescent imaging, we were able to follow pMM cells outgrowth and to visualize the effect of treatment. Therapeutic interventions in this model resulted in equivalent drug responses as observed in the corresponding patients. This novel human-mouse hybrid model creates unprecedented opportunities to investigate species-specific microenvironmental influences on normal and malignant hematopoietic development, and to develop and personalize cancer treatment strategies.
- Published
- 2012
- Full Text
- View/download PDF
49. Redirecting αβT cells against cancer cells by transfer of a broadly tumor-reactive γδT-cell receptor
- Author
-
Marcu-Malina, Victoria, Heijhuurs, Sabine, van Buuren, Marit, Hartkamp, Linda, Strand, Susanne, Sebestyen, Zsolt, Scholten, Kirsten, Martens, Anton, and Kuball, Jürgen
- Abstract
Major limitations of currently investigated αβT cells redirected against cancer by transfer of tumor-specific αβTCR arise from their low affinity, MHC restriction, and risk to mediate self-reactivity after pairing with endogenous α or βTCR chains. Therefore, the ability of a defined γ9δ2TCR to redirect αβT cells selectively against tumor cells was tested and its molecular interaction with a variety of targets investigated. Functional analysis revealed that a γ9δ2TCR efficiently reprograms both CD4+and CD8+αβT cells against a broad panel of cancer cells while ignoring normal cells, and substantially reduces but does not completely abrogate alloreactivity. γ9δ2TCR-transduced αβT cells reduced colony formation of progenitor cells of primary acute myeloid leukemia blasts and inhibited leukemia growth in a humanized mouse model. Thereby, metabolites of a dysregulated mevalonate pathway are targeted and the additional application of widely used biphosphonates is crucial for in vivo efficacy most likely because of its modulating effect on cytokine secretion of γ9δ2TCR-transduced αβT cells. Expression of NKG2D ligands and F1-ATPase contributed to the activity of γ9δ2TCR-transduced αβT cells but were not mandatory. In summary, γ9δ2 TCRs are an attractive alternative to broadly redirect αβT cells against cancer cells with both an improved efficacy and safety profile compared with currently used αβTCRs.
- Published
- 2011
- Full Text
- View/download PDF
50. Redirecting αβT cells against cancer cells by transfer of a broadly tumor-reactive γδT-cell receptor
- Author
-
Marcu-Malina, Victoria, Heijhuurs, Sabine, van Buuren, Marit, Hartkamp, Linda, Strand, Susanne, Sebestyen, Zsolt, Scholten, Kirsten, Martens, Anton, and Kuball, Jürgen
- Abstract
Major limitations of currently investigated αβT cells redirected against cancer by transfer of tumor-specific αβTCR arise from their low affinity, MHC restriction, and risk to mediate self-reactivity after pairing with endogenous α or βTCR chains. Therefore, the ability of a defined γ9δ2TCR to redirect αβT cells selectively against tumor cells was tested and its molecular interaction with a variety of targets investigated. Functional analysis revealed that a γ9δ2TCR efficiently reprograms both CD4+ and CD8+ αβT cells against a broad panel of cancer cells while ignoring normal cells, and substantially reduces but does not completely abrogate alloreactivity. γ9δ2TCR-transduced αβT cells reduced colony formation of progenitor cells of primary acute myeloid leukemia blasts and inhibited leukemia growth in a humanized mouse model. Thereby, metabolites of a dysregulated mevalonate pathway are targeted and the additional application of widely used biphosphonates is crucial for in vivo efficacy most likely because of its modulating effect on cytokine secretion of γ9δ2TCR-transduced αβT cells. Expression of NKG2D ligands and F1-ATPase contributed to the activity of γ9δ2TCR-transduced αβT cells but were not mandatory. In summary, γ9δ2 TCRs are an attractive alternative to broadly redirect αβT cells against cancer cells with both an improved efficacy and safety profile compared with currently used αβTCRs.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.