106 results on '"Leemhuis T"'
Search Results
2. Essential requirements for setting up a stem cell processing laboratory
- Author
-
Leemhuis, T, Padley, D, Keever-Taylor, C, Niederwieser, D, Teshima, T, Lanza, F, Chabannon, C, Szabolcs, P, Bazarbachi, A, and Koh, M B C
- Published
- 2014
- Full Text
- View/download PDF
3. Transplantation of a fetus with paternal Thy-1+CD34+cells for chronic granulomatous disease
- Author
-
Muench, MO, Rae, J, Bárcena, A, Leemhuis, T, Farrell, J, Humeau, L, Maxwell-Wiggins, JR, Capper, J, Mychaliska, GB, Albanese, CT, Martin, T, Tsukamoto, A, Curnutte, JT, and Harrison, MR
- Published
- 2001
- Full Text
- View/download PDF
4. Post-Thaw Viability of HPC Products Is Unaffected by Minor Freezing Curve Variations with the Use of 2.5% HES and 5% DMSO in the Cryopreservation Media: SP440
- Author
-
Schuesler, T, Bruns, C, Petrola, F, Lamping, J, and Leemhuis, T
- Published
- 2010
5. Isolation of Hematopoietic Progenitor Cells from Peripheral Blood of Sickle Cell Disease Patients: A Feasibility Study: SP426
- Author
-
Bruns, C, Perumbeti, A, Temples, T, Grassman, E, Cancelas, J A, Malik, P, and Leemhuis, T
- Published
- 2010
6. Treatment of Ligneous Conjunctivitis with Topical Administration of Allogeneic Fresh Frozen Plasma: S59-030D
- Author
-
Leemhuis, T, Carey, P M, Bush, S, Hodgson, C, and Palumbo, J S
- Published
- 2010
7. Red Blood Cell Depletion of Allogeneic Bone Marrow Using Hetastarch: SP49
- Author
-
Griggs, P, Bruns, C, and Leemhuis, T
- Published
- 2007
8. Clinical-Scale Transduction of a Drug Resistance Gene into Hematopoietic Progenitor Cells Using a Retroviral Vector: S68–040A
- Author
-
Balcik, B, Leemhuis, T, Murphy, K, Reeves, L, Wagner, L, and Williams, D
- Published
- 2007
9. Improved Post Thaw Viability And Engraftment Kinetics After Reducing The DMSO Concentration And Adding Hetastartch To The Cryoprotectant Solution Used To Cryopreserve Hematopoietic Cell Populations: S21-030E
- Author
-
Balcik, B, Griggs, P, Inglish, P, and Leemhuis, T
- Published
- 2005
10. Production of quadrivalent virus-specific t cells utilizing peptide stimulation
- Author
-
Damen, A., primary, Heyenbruch, D., additional, Gong, N., additional, Maurer, K., additional, Grimley, M., additional, Nelson, A., additional, Davies, S., additional, Lutzko, C., additional, Zhu, X., additional, Bollard, C., additional, Hanley, P., additional, and Leemhuis, T., additional
- Published
- 2018
- Full Text
- View/download PDF
11. TCR α/β+ T cell and CD19+ B cell depletion of HPC apheresis products utilizing CliniMACS
- Author
-
Frondorf, M., primary, Brummer, J., additional, Gul, Z., additional, and Leemhuis, T., additional
- Published
- 2018
- Full Text
- View/download PDF
12. 294 - Production of quadrivalent virus-specific t cells utilizing peptide stimulation
- Author
-
Damen, A., Heyenbruch, D., Gong, N., Maurer, K., Grimley, M., Nelson, A., Davies, S., Lutzko, C., Zhu, X., Bollard, C., Hanley, P., and Leemhuis, T.
- Published
- 2018
- Full Text
- View/download PDF
13. 178 - TCR α/β+ T cell and CD19+ B cell depletion of HPC apheresis products utilizing CliniMACS
- Author
-
Frondorf, M., Brummer, J., Gul, Z., and Leemhuis, T.
- Published
- 2018
- Full Text
- View/download PDF
14. Persistence of human multilineage, self-renewing lymphohematopoietic stem cells in chimeric sheep
- Author
-
Ef, Srour, Ed, Zanjani, Kenneth Cornetta, Cm, Traycoff, Aw, Flake, Hedrick M, Je, Brandt, Leemhuis T, and Hoffman R
- Subjects
Sheep ,Base Sequence ,Chimera ,Molecular Sequence Data ,Transplantation, Heterologous ,Immunology ,Antigens, CD34 ,Bone Marrow Cells ,HLA-DR Antigens ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Biochemistry ,Hematopoiesis ,Antigens, CD ,Pregnancy ,Animals ,Humans ,Female ,Lymphocytes ,Bone Marrow Transplantation - Abstract
We have previously reported the ability of uncharacterized human bone marrow (BM) cells to engraft into preimmune fetal sheep, thereby creating sheep-human chimera suitable for in vivo examination of the properties of human hematopoietic stem cells (HSC). Adult human bone marrow CD34+ HLA-DR- cells have been extensively characterized in vitro and have been demonstrated to contain a number of primitive hematopoietic progenitor cells (PHPC). However, the capacity of such highly purified populations of human marrow CD34+ HLA-DR- cells to undergo in vivo self-renewal and multipotential lymphohematopoietic differentiation has not been previously demonstrated. To achieve that, human CD34+ HLA-DR- cells were transplanted in utero into immunoincompetent fetal sheep to investigate the BM-populating potential of these cells. Long-term chimerism, sustained human hematopoiesis, and expression of human cells belonging to all human blood cell lineages were demonstrated in two animals for more than 7 months' posttransplantation. Chimeric BM contained erythroid, granulocytic/monocytic, and megakaryocytic hematopoietic progenitor cells, as well as the primitive high proliferative potential colony- forming cell (HPP-CFC). Under a variety of in vitro experimental conditions, chimeric BM cells gave rise to human T cells expressing T- lymphocyte-specific markers, human natural killer (NK) cells, and human IgG-producing B cells. In vivo expansion and possibly self-renewal of transplanted PHPC was confirmed by the detection in chimeric BM 130 days' posttransplantation of CD34+ HLA-DR- cells, the phenotype of human cells constituting the stem-cell graft. These studies demonstrate not only the BM-populating capacity, multipotential differentiation, and most likely self-renewal capabilities of human CD34+ HLA-DR- cells, but also that this BM population contains human HSC. Furthermore, it appears that this animal model of xenogeneic stem-cell transplantation is extremely useful for in vivo examination of human hematopoiesis and the behavioral and functional characteristics of human HSC.
- Published
- 1993
- Full Text
- View/download PDF
15. Simultaneous mobilization of Mac-1 (CD11b/CD18) and formyl peptide chemoattractant receptors in human neutrophils [published erratum appears in Blood 1993 Mar 15;81(6):1668]
- Author
-
EF Strour, Leo J. McCarthy, Gabig T, Denis English, Graves, and Leemhuis T
- Subjects
medicine.medical_treatment ,Immunology ,Cell ,Chemotaxis ,Inflammation ,Cell Biology ,Hematology ,Biochemistry ,Exocytosis ,Cell biology ,chemistry.chemical_compound ,Cytokine ,medicine.anatomical_structure ,chemistry ,Specific granule ,Downregulation and upregulation ,medicine ,medicine.symptom ,Receptor ,Fluorescein isothiocyanate ,Intracellular - Abstract
Mobilization of a distinct subset of specific granules provides a physiologically important mechanism to recruit Mac-1 (CD11b/CD18) from an intracellular pool to the external surface of the neutrophil plasma membrane, where the functionally active heterodimer mediates several adherence-dependent processes that are crucial for adequate host defense and cellular inflammatory responses. We observed similar characteristics for translocation of Mac-1 and neutrophil formyl peptide receptors (FPR) and hypothesize that the readily accessible pools of both Mac-1 and FPR are colocal-ized within this specific granule subset. Plasma membrane levels of both FPR (assessed with 3 H-FMLP) and Mac-1 (assessed by fluorescence-activated cell sorter analysis of fluorescein isothiocyanate [FITC]-Mo-1-labeled cells) were markedly downregulated in cells prepared at low temperature from blood cooled to 4°C immediately after removal from the circulation. Levels of both FPR and Mac-1 remained low on cells held at 4°C. Upon warming, spontaneous upreg-ulation of Mac-1 and FPR occurred with similar kinetics and temperature dependency. Translocation of both Mac-1 and FPR was markedly potentiated by exposure of cells to either fluoride ion (which has been shown by others to specifically elicit exocytosis of gelatinase-rich and vitamin B-12 binding protein-poor granules) or granulocyte-macrophage colony-stimulating factor (GM-CSF), a cytokine that markedly potentiates the neutrophils’ host defense capabilities. Levels of both FPR and Mac-1 on F- or GM-CSF-treated neutrophils exceeded those present on cells incubated at 37°C for extended time intervals, indicating that stimulated translocation may involve mobilization of an additional granule sub-set. Scatchard analysis showed that only low-affinity FPR were translocated during spontaneous and stimulus-depen-dent upregulation. To directly compare FPR levels on the surface of cells displaying varying levels of Mac-1 within a single cell suspension, cells were labeled with FITC-Mo-1 and sorted into subpopulations based on fluorescence intensity. After sorting, the individual populations were held at 4°C to prevent further spontaneous upregulation, concentrated by centrifugation, and assayed for FPR levels. Under a variety of conditions, FPR levels correlated with Mac-1 (CD11b) expression on cell populations selected on the basis of CD11b fluorescence intensity. Analysis of subcellular fractions obtained from disrupted neutrophils before and after upregula-tion provided additional support for the hypothesis that Mac-1 and FPR are colocalized within a readily accessible subset of neutrophil granules. After the cells marginate and leave the circulation in response to signals recognized by receptors for formyl peptides or other chemoattractants and cellular agonists, simultaneous translocation of Mac-1 and low-affinity FPR from tertiary granules to the plasma membrane provides neutrophils with an efficient mechanism to amplify multiple aspects of their inflammatory and host defense capabilities. Complex interactions between newly deployed receptors and adherence-related antigens with signal-transducing G-proteins, phospholipases, and other membrane proteins may convey crucial regulatory influences on the responsiveness of neutrophils at sites of infection and inflammation. © 1992 by The American Society of Hematology. 0006-4971/92/8003-0019$3.00/0
- Published
- 1992
- Full Text
- View/download PDF
16. Analysis of HPC and HSC post-thaw viability, clonogenic capacity, and proliferative potential after cryopreservation in comercially available media
- Author
-
Almulhem, N., primary, Lutzko, C., additional, and Leemhuis, T., additional
- Published
- 2013
- Full Text
- View/download PDF
17. Large volume whole blood mononuclear cell enrichment using a COBE spectra
- Author
-
Almezel, N., primary, Pinkard, S., additional, and Leemhuis, T., additional
- Published
- 2013
- Full Text
- View/download PDF
18. Characterization of normal human CD3+ CD5- and γδ T cell receptor positive T lymphocytes
- Author
-
SROUR, E. F., primary, LEEMHUIS, T., additional, JENSKI, L., additional, REDMOND, R., additional, FILLAK, D., additional, and JANSEN, J., additional
- Published
- 2008
- Full Text
- View/download PDF
19. 682. Collection and Genetic Correction of Fanconi Complement Type A Hematopoietic Stem Cells: Results of Two Pilot Studies
- Author
-
Kelly, P.F., primary, Balcik, B., additional, Bohn, K., additional, Mueller, R., additional, Schuesler, T., additional, Reeves, L., additional, Leemhuis, T., additional, Harris, R., additional, Davies, S.M., additional, Smith, F.O., additional, von Kalle, C., additional, and Williams, D.A., additional
- Published
- 2006
- Full Text
- View/download PDF
20. Early Results of the Clinical Collection and Genetic Correction of Fanconi Complement Type A Hematopoietic Stem Cells.
- Author
-
Kelly, Patrick, primary, Balcik, B., primary, Bohn, K., primary, Mueller, R., primary, Jurickova, I., primary, Schuesler, T., primary, Reeves, L., primary, Leemhuis, T., primary, Harris, R., primary, Davies, S. M., primary, Smith, F. O., primary, von Kalle, C., primary, and Williams, D. A., primary
- Published
- 2005
- Full Text
- View/download PDF
21. Collection, Tumor Contamination, and Engraftment Kinetics of Highly Purified Hematopoietic Progenitor Cells to Support High Dose Therapy in Multiple Myeloma
- Author
-
Tricot, G., primary, Gazitt, Y., additional, Leemhuis, T., additional, Jagannath, S., additional, Desikan, K.R., additional, Siegel, D., additional, Fassas, A., additional, Tindle, S., additional, Nelson, J., additional, Juttner, C., additional, Tsukamoto, A., additional, Hallagan, J., additional, Atkinson, K., additional, Reading, C., additional, Hoffman, R., additional, and Barlogie, B., additional
- Published
- 1998
- Full Text
- View/download PDF
22. Persistence of human multilineage, self-renewing lymphohematopoietic stem cells in chimeric sheep
- Author
-
Srour, EF, primary, Zanjani, ED, additional, Cornetta, K, additional, Traycoff, CM, additional, Flake, AW, additional, Hedrick, M, additional, Brandt, JE, additional, Leemhuis, T, additional, and Hoffman, R, additional
- Published
- 1993
- Full Text
- View/download PDF
23. Long-term generation and expansion of human primitive hematopoietic progenitor cells in vitro
- Author
-
Srour, EF, primary, Brandt, JE, additional, Briddell, RA, additional, Grigsby, S, additional, Leemhuis, T, additional, and Hoffman, R, additional
- Published
- 1993
- Full Text
- View/download PDF
24. Identification of BCR/ABL-negative primitive hematopoietic progenitor cells within chronic myeloid leukemia marrow
- Author
-
Leemhuis, T, primary, Leibowitz, D, additional, Cox, G, additional, Silver, R, additional, Srour, EF, additional, Tricot, G, additional, and Hoffman, R, additional
- Published
- 1993
- Full Text
- View/download PDF
25. Simultaneous mobilization of Mac-1 (CD11b/CD18) and formyl peptide chemoattractant receptors in human neutrophils [published erratum appears in Blood 1993 Mar 15;81(6):1668]
- Author
-
Graves, V, primary, Gabig, T, additional, McCarthy, L, additional, Strour, EF, additional, Leemhuis, T, additional, and English, D, additional
- Published
- 1992
- Full Text
- View/download PDF
26. Sustained human hematopoiesis in sheep transplanted in utero during early gestation with fractionated adult human bone marrow cells
- Author
-
Srour, EF, primary, Zanjani, ED, additional, Brandt, JE, additional, Leemhuis, T, additional, Briddell, RA, additional, Heerema, NA, additional, and Hoffman, R, additional
- Published
- 1992
- Full Text
- View/download PDF
27. Relationship between cytokine-dependent cell cycle progression and MHC class II antigen expression by human CD34+ HLA-DR- bone marrow cells.
- Author
-
Srour, E F, primary, Brandt, J E, additional, Leemhuis, T, additional, Ballas, C B, additional, and Hoffman, R, additional
- Published
- 1992
- Full Text
- View/download PDF
28. Transplantation of a fetus with paternal Thy-1+CD34+cells for chronic granulomatous disease.
- Author
-
Muench, M O, Rae, J, Bárcena, A, Leemhuis, T, Farrell, J, Humeau, L, Maxwell-Wiggins, J R, Capper, J, Mychaliska, G B, Albanese, C T, Martin, T, Tsukamoto, A, Curnutte, J T, and Harrison, M R
- Subjects
FETUS ,CHRONIC granulomatous disease ,STEM cells - Abstract
A fetus diagnosed with X-linked chronic granulomatous disease was transplanted with Thy-1
+ CD34+ cells of paternal origin. The transplant was performed at 14 weeks gestation by ultrasound guided injection into the peritoneal cavity. The fetus was delivered at 38 weeks gestation after an otherwise uneventful pregnancy. Umbilical cord blood was collected and used to determine the level of peripheral blood chimerism as well as levels of functional engrafted cells. Flow cytometry was used to detect donor leukocytes identified as HLA-A2- B7+ cells, whereas recipient cells were identified as HLA-A2+ B7- cells. No evidence of donor cell engraftment above a level of 0.01% was found. PCR was used to detect HLA-DRB1*15+ donor cells among the recipient’s HLA-DRB1*15- cells, but no engraftment was seen with a sensitivity of 1:1000. The presence of functional, donor-derived neutrophils was assessed by flow cytometry using two different fluorescent dyes that measure reactive oxygen species generated by the phagocyte NADPH oxidase. No evidence of paternal-derived functional neutrophils above a level of 0.15% was observed. Peripheral blood and bone marrow samples were collected at 6 months of age. Neither sample showed engraftment by HLA typing using both flow cytometry and PCR. Functional phagocytes were also not observed. Furthermore, no indication of immunological tolerance specific for the donor cells was indicated by a mixed lymphocyte reaction assay performed at 6 months of age. While there appears to be no engraftment of the donor stem cells, the transplant caused no harm to the fetus and the child was healthy at 6 months of age. Analyses of fetal tissues, obtained from elective abortions, revealed that CD3+ T cells and CD56+ CD3- NK cells are present in the liver at 8 weeks gestation and in the blood by 9 weeks gestation. The presence of these... [ABSTRACT FROM AUTHOR]- Published
- 2001
- Full Text
- View/download PDF
29. Characterization of normal human CD3+ CD5- and γδ T cell receptor positive T lymphocytes.
- Author
-
Srour, E. F., Leemhuis, T., Jenski, L., Redmond, R., Fillak, D., and Jansen, J.
- Subjects
- *
T cell receptors , *LYMPHOCYTES , *LEUCOCYTES , *IMMUNOFLUORESCENCE , *CELL membranes , *IMMUNOGLOBULINS - Abstract
The functional and phenotypic properties of normal human CD3+CD5- T cells which have a higher frequency of cytotoxic cells than CD3+CD5+ T lymphocytes have been described. Using three- and four-colour immunofluorescence flow cytometric cell sorting, the CD3+CD5- and CD3+CD5+ populations were subdivided into αβ or γδ T cell receptor positive cells. The four subsets were examined for the in vitro cytotoxic activity and were also stimulated with mitogens in limiting-dilution assays to measure the frequencies of proliferating and interleukin-2 (IL-2) producing cells. CD+CD5-αβ+, CD3+CD5-γδ+ and CD3+CD5+γδ+ cells had lower frequencies of proliferating and IL-2-producing cells than did CD3+CD5+αβ+ cells. However, the cytotoxic activity of the different phenotypes was higher in the CD3+CD5- subsets, especially when these cells were γδ+. Expression of γδ or lack of expression of CD5 appeared to be associated with the acquisition of cytolytic potentials. CD8 was expressed on 20% of fresh CD3+γδ+ cells. Cultured γδ+ cells retained the expression of γδ, but quickly lost that of CD8 and with time modulated the expression of CD5. The expression of CD5 was found to be higher on sorted CD3+CD5+γδ- than on CD3+CD5-γδ+ cells. These observations indicate that γδ is preferentially expressed on CD5-negative or weakly positive T lymphocytes and that CD3+CD5-γδ+ cells appear to constitute a discrete small subset of mature T lymphocytes which are cytotoxic in nature. However, the exact immunological function of these cells and their place in T cell ontogeny are yet to be elucidated. [ABSTRACT FROM AUTHOR]
- Published
- 1990
- Full Text
- View/download PDF
30. Effects of recombinant human colony stimulating factors (CSF) (granulocyte-macrophage CSF, granulocyte CSF, and CSF-1) on human monocyte/macrophage differentiation
- Author
-
klaus geissler, Harrington M, Srivastava C, Leemhuis T, Tricot G, and He, Broxmeyer
- Subjects
Naphthol AS D Esterase ,Macrophages ,Nitroblue Tetrazolium ,Immunology ,Granulocyte-Macrophage Colony-Stimulating Factor ,Cell Differentiation ,Receptor, Macrophage Colony-Stimulating Factor ,Proto-Oncogene Mas ,Monocytes ,Recombinant Proteins ,Cell Line ,Colony-Stimulating Factors ,Proto-Oncogene Proteins ,Antigens, Surface ,Granulocyte Colony-Stimulating Factor ,Humans ,Immunology and Allergy ,Growth Substances ,Oxidation-Reduction ,Cell Division ,Granulocytes - Abstract
Purified recombinant human granulocyte-macrophage (rhuGM)-CSF, rhuG-CSF, and rhuCSF-1 were evaluated for their capacity to influence the differentiation of U-937 cells and normal human monocytes. The human U-937 cell line represents an early stage of monocytic differentiation. It was found that rhuGM-CSF and rhuG-CSF, but not rhuCSF-1, induced phenotypic changes consistent with monocyte/macrophage differentiation in U-937 cells. After 3 days of culture in the presence of either rhuGM-CSF or rhuG-CSF, a small but significant proportion of U-937 cells were able to reduce nitroblue tetrazolium. Nitroblue tetrazolium reduction, however, was maximally induced when rhuGM-CSF and rhuG-CSF were added in combination. These changes were accompanied by increased alpha-naphthyl acetate esterase activity, acquisition of macrophage morphology, Mo-1 Ag expression, and decreased cell proliferation. rhuGM-CSF alone also induced expression of the c-fms proto-oncogene (CSF-1 receptor) in U-937 cells and this expression was enhanced by the combination of rhuGM-CSF and rhuG-CSF. In cultured normal human peripheral blood monocytes, representing a late stage of maturation, rhuGM-CSF and rhuCSF-1 differentially increased Mo-1 and My-4 Ag expression, respectively, whereas rhuG-CSF was without effect. Our results suggest that the interaction of GM-CSF, G-CSF, and CSF-1 may play a fundamental role in the early and late stages of the human monocyte/macrophage differentiation process.
- Published
- 1989
- Full Text
- View/download PDF
31. Differentiation-inducing effect of recombinant human tumor necrosis factor alpha and gamma-interferon in vitro on blast cells from patients with acute myeloid leukemia and myeloid blast crisis of chronic myeloid leukemia
- Author
-
klaus geissler, Tricot G, Leemhuis T, Walker E, and He, Broxmeyer
- Subjects
Adult ,Male ,Adolescent ,Tumor Necrosis Factor-alpha ,Cell Differentiation ,Middle Aged ,Recombinant Proteins ,Interferon-gamma ,Leukemia, Myeloid, Acute ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Cell Adhesion ,Humans ,Female ,Blast Crisis ,Cell Division ,Aged - Abstract
Tumor necrosis factor alpha (TNF-alpha) and gamma-interferon (IFN-gamma) have been shown to suppress clonogenic growth in cultures containing blast cells obtained from patients with acute myeloid leukemia. We report that recombinant human TNF-alpha and IFN-gamma are also able to induce functional and morphological maturation in fresh myeloid leukemic cells in vitro. Assessing suspension cultures containing cells from patients with acute myeloid leukemia (11 patients) or myeloid blast crisis of chronic myeloid leukemia (5 patients), it was found that recombinant human TNF-alpha and IFN-gamma significantly enhanced the number of cells reducing nitroblue tetrazolium, as compared to control cultures containing no cytokine (P less than 0.001 and P less than 0.001, respectively). Cells from responders showed alterations characteristic of monocyte/macrophage differentiation, adherence to plastic surfaces, development of positive staining for alpha-naphthyl acetate esterase, typical morphology, and expression of cell surface antigens detected by the monoclonal antibodies Mo-1, Mo-2, and My-4. Both cytokines decreased the number of viable cells, the number of blast cells, and the number of cluster-forming units in suspension culture, suggesting inhibitory actions on the growth capacity of leukemic cells. Compared to the maximum effects of either factor alone, the combination of recombinant human TNF-alpha and IFN-gamma significantly increased the extent of growth inhibition and cell adherence but did not result in further increases in nitroblue tetrazolium reduction. The presence of Auer rods in IFN-gamma or TNF-alpha differentiation-induced macrophages with cells from a patient with M5 acute myeloid leukemia demonstrates that these cytokines can induce differentiation of a leukemic clone in primary cells from patients with leukemia.
32. Effects of recombinant human colony stimulating factors (CSF) (granulocyte-macrophage CSF, granulocyte CSF, and CSF-1) on human monocyte/macrophage differentiation.
- Author
-
Geissler, K, primary, Harrington, M, additional, Srivastava, C, additional, Leemhuis, T, additional, Tricot, G, additional, and Broxmeyer, H E, additional
- Published
- 1989
- Full Text
- View/download PDF
33. Murine B lymphoma cell lines release functionally active interleukin 2 after stimulation with Staphylococcus aureus.
- Author
-
Walker, E, primary, Leemhuis, T, additional, and Roeder, W, additional
- Published
- 1988
- Full Text
- View/download PDF
34. Third-party virus-specific T cells for the treatment of double-stranded DNA viral reactivation and posttransplant lymphoproliferative disease after solid organ transplant.
- Author
-
Khoury R, Grimley MS, Nelson AS, Leemhuis T, Cancelas JA, Cook E, Wang Y, Heyenbruch D, Bollard CM, Keller MD, Hanley PJ, Lutzko C, Pham G, Davies SM, and Rubinstein JD
- Subjects
- Humans, Male, Middle Aged, Female, Adult, Postoperative Complications, DNA, Viral, Aged, Cytomegalovirus, Prognosis, Follow-Up Studies, Herpesvirus 4, Human, Young Adult, DNA Virus Infections virology, Organ Transplantation adverse effects, Lymphoproliferative Disorders etiology, Lymphoproliferative Disorders virology, Lymphoproliferative Disorders therapy, T-Lymphocytes immunology, Virus Activation
- Abstract
Reactivation or primary infection with double-stranded DNA viruses is common in recipients of solid organ transplants (SOTs) and is associated with significant morbidity and mortality. Treatment with conventional antiviral medications is limited by toxicities, resistance, and a lack of effective options for adenovirus (ADV) and BK polyomavirus (BKPyV). Virus-specific T cells (VSTs) have been shown to be an effective treatment for infections with ADV, BKPyV, cytomegalovirus (CMV), and Epstein-Barr virus (EBV). Most of these studies have been conducted in stem cell recipients, and no large studies have been published in the SOT population to date. In this study, we report on the outcome of quadrivalent third-party VST infusions in 98 recipients of SOTs in the context of an open-label phase 2 trial. The 98 patients received a total of 181 infusions, with a median of 2 infusions per patient. The overall response rate was 45% for BKPyV, 65% for cytomegalovirus, 68% for ADV, and 61% for Epstein-Barr virus. Twenty percent of patients with posttransplant lymphoproliferative disorder had a complete response and 40% of patients had a partial response. All the VST infusions were well tolerated. We conclude that VSTs are safe and effective in the treatment of viral infections in SOT recipients., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
35. Third-Party and Patient-Specific Donor-Derived Virus-Specific T Cells Demonstrate Similar Efficacy and Safety for Management of Viral Infections after Hematopoietic Stem Cell Transplantation in Children and Young Adults.
- Author
-
Galletta TJ, Lane A, Lutzko C, Leemhuis T, Cancelas JA, Khoury R, Wang YM, Hanley PJ, Keller MD, Bollard CM, Davies SM, Grimley MS, and Rubinstein JD
- Subjects
- Child, Humans, Young Adult, Herpesvirus 4, Human, Retrospective Studies, T-Lymphocytes, Transplantation, Homologous, Epstein-Barr Virus Infections, Hematopoietic Stem Cell Transplantation adverse effects, Virus Diseases etiology, Virus Diseases therapy
- Abstract
Infections with double-stranded DNA viruses are a common complication after hematopoietic stem cell transplantation (HSCT) and cause significant morbidity and mortality in the post-transplantation period. Both donor-derived (DD) and third-party (TP) virus-specific T cells (VSTs) have shown efficacy and safety in viral management following HSCT in children and young adults. Owing to a greater degree of HLA matching between the recipient and stem cell donor, DD VSTs potentially persist longer in circulation compared to TP VSTs, because they are collected from a well-matched donor. However, TP VSTs are more easily accessible, particularly for smaller transplantation centers that do not have VST manufacturing capabilities, and more economical than creating a customized product for each transplant recipient. We conducted the present study to compare clinical efficacy and safety outcomes for DD VSTs and TP VSTs in a large cohort of pediatric and young adult HSCT recipients and to determine whether DD VSTs are associated with improved outcomes owing to potentially longer persistence in the recipient's circulation. This retrospective cohort study included 145 patients who received VSTs at Cincinnati Children's Hospital Medical Center (CCHMC) between 2017 and 2021 for the treatment of adenovirus, BK virus, cytomegalovirus, and/or Epstein-Barr virus. Viruses were detected using quantitative polymerase chain reaction. Patients received VSTs on a DD (NCT02048332) or TP (NCT02532452) protocol, and VST products for both protocols were manufactured in an identical fashion. The primary study outcome was clinical response to VSTs, evaluated 4 weeks after VST administration, defined as decrease in viral load to under the inclusion thresholds, or resolution of symptoms of invasive viral infection, without the need for additional conventional antiviral medication following VST administration. Secondary outcomes included graft-versus-host-disease, transplant-associated thrombotic microangiopathy, renal function, hospital length of stay, and overall survival at 30 days and 100 days after VST administration and 1 year after HSCT. Statistical analysis was performed using the Fisher exact test or chi-square test. An unpaired t test was used to compare continuous variables. The study group comprised 77 patients in the DD cohort and 68 patients in the TP cohort. Eighteen patients in the TP cohort underwent HSCT at CCHMC, and the other 50 underwent HSCT at other institutions and presented to CCHMC solely for VST administration. There was no statistically significant difference in clinical response rates between DD and TP cohorts (65.6% versus 62.7%; odds ratio [OR], 1.162; 95% confidence interval [CI], .619 to 2.164; P = .747). There were no significant differences in secondary outcomes between the 2 cohorts. The percentage of patients requiring multiple infusions for a clinical response did not differ significantly between the DD and TP cohorts (38.2% versus 32.5%; OR, .780; 95% CI, .345 to 1.805; P = .666). We found no significant difference in clinical response rate between DD VSTs and TP VSTs and a similar safety profile. Our data suggest that TP VSTs may be sufficient to control viral infection until immune reconstitution occurs despite the potential for more rapid VST clearance compared to DD VSTs. The lack of significant differences between DD VSTs and TP VSTs is an important finding, indicating that it is not necessary for every transplant center to manufacture customized DD VSTs, and that TP VSTs are a satisfactory substitute., (Copyright © 2023 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
36. Scheduled administration of virus-specific T cells for viral prophylaxis after pediatric allogeneic stem cell transplant.
- Author
-
Rubinstein JD, Lutzko C, Leemhuis T, Zhu X, Pham G, Ray L, Thomas S, Dourson C, Wilhelm J, Lane A, Cancelas JA, Lipps D, Ferrell J, Hanley PJ, Keller MD, Bollard CM, Wang YM, Davies SM, Nelson AS, and Grimley MS
- Subjects
- Child, Herpesvirus 4, Human, Humans, T-Lymphocytes, Viremia etiology, Epstein-Barr Virus Infections, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
Infections with double-stranded DNA viruses are a significant cause of morbidity and mortality in pediatric patients following allogeneic hematopoietic stem cell transplantation (HSCT). Virus-specific T-cell therapies (VSTs) have been shown to be an effective treatment for infections with adenovirus, BK virus, cytomegalovirus (CMV), and Epstein-Barr virus (EBV). To date, prophylactic regimens to prevent or mitigate these infections using conventional antiviral medications provide suboptimal response rates. Here we report on a clinical trial (NCT03883906) performed to assess the feasibility of rapid manufacturing and early infusion of quadrivalent VSTs generated from stem cell donors ("donor-derived VSTs") into allogeneic HSCT recipients with minimal or absent viremia. Patients were eligible to receive scheduled VSTs as early as 21 days after stem cell infusion. Twenty-three patients received scheduled VSTs. Twenty of 23 patients had no viremia at the time of infusion, while 3 patients had very low-level BK viremia. Two developed clinically significant graft-versus-host disease (GVHD), although this incidence was not outside of expected incidence early after HSCT, and both were successfully treated with systemic corticosteroids (n = 2). Five patients were deemed treatment failures. Three developed subsequent significant viremia/viral disease (n = 3). Eighteen patients did not fail treatment, 7 of whom did not develop any viremia, while 11 developed low-level, self-limited viremia that resolved without further intervention. No infusion reactions occurred. In conclusion, scheduled VSTs appear to be safe and potentially effective at limiting serious complications from viral infections after allogeneic transplantation. A randomized study comparing this scheduled approach to the use of VSTs to treat active viremia is ongoing., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
37. Off-the-Shelf Third-Party Virus-Specific T Cell Therapy to Treat JC Polyomavirus Infection in Hematopoietic Stem Cell Transplantation Recipients.
- Author
-
Rubinstein JD, Jodele S, Heyenbruch D, Wilhelm J, Thomas S, Lutzko C, Zhu X, Leemhuis T, Cancelas JA, Keller M, Bollard CM, Hanley PJ, Boghdadly ZE, Mims A, Davies SM, Grimley MS, and Nelson AS
- Subjects
- Cell- and Tissue-Based Therapy, Child, Humans, Retrospective Studies, Hematopoietic Stem Cell Transplantation adverse effects, JC Virus, Leukoencephalopathy, Progressive Multifocal etiology, Polyomavirus Infections therapy
- Abstract
Progressive multifocal leukoencephalopathy (PML) is a progressive and generally fatal demyelinating neurologic disease that occurs in profoundly immunocompromised patients due to infection with the human polyomavirus JC virus (JCPyV). Treatment options are limited and are largely focused on restoring T cell immunity, and outcomes are historically poor. Control of JCPyV in the setting of an immunocompromised patient by adoptive transfer of third-party virus specific T cells (VSTs) has been described in a small number of cases. To investigate treatment response and outcomes in recipients of hematopoietic stem cell transplantation (HSCT) with PML treated with third-party VSTs directed against the BK virus, a highly homologous polyoma virus that shares immunogenic epitopes with JCPyV. A retrospective chart review was performed on 4 patients who received VSTs for the treatment of PML at Cincinnati Children's Hospital Medical Center since 2019. VSTs were administered safely, with no cases of graft-versus-host disease and no infusion reactions. One patient who was treated almost immediately after diagnosis was able to clear JCPyV from blood and cerebrospinal fluid, with resultant stabilization of neurologic decline. IFN-γ enzyme-linked immunospot (ELISpot) analysis demonstrated VSTs in the peripheral blood following infusion. Response was maintained through repeat infusions. Three other patients, all of whom had a longer delay between diagnosis and infusion, exhibited progressive neurologic decline despite varying degrees of improvement in viral load. PML is a rare but often fatal complication following HSCT for which few treatment options are available. BK-directed, JCPyV cross-reactive VSTs are a safe and viable therapeutic option, and prompt administration should be considered once PML is diagnosed. © 2021 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc., (Copyright © 2021 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
38. Virus-specific T cells for adenovirus infection after stem cell transplantation are highly effective and class II HLA restricted.
- Author
-
Rubinstein JD, Zhu X, Leemhuis T, Pham G, Ray L, Emberesh S, Jodele S, Thomas S, Cancelas JA, Bollard CM, Hanley PJ, Keller MD, Grimley O, Clark D, Clark T, Lindestam Arlehamn CS, Sette A, Davies SM, Nelson AS, Grimley MS, and Lutzko C
- Subjects
- Child, Humans, Interferon-gamma, Stem Cell Transplantation adverse effects, T-Lymphocytes, Adenoviridae Infections therapy, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
Infection with adenoviruses is a common and significant complication in pediatric patients after allogeneic hematopoietic stem cell transplantation. Treatment options with traditional antivirals are limited by poor efficacy and significant toxicities. T-cell reconstitution is critical for the management of adenoviral infections, but it generally takes place months after transplantation. Ex vivo-generated virus-specific T cells (VSTs) are an alternative approach for viral control and can be rapidly generated from either a stem cell donor or a healthy third-party donor. In the context of a single-center phase 1/2 clinical trial, we treated 30 patients with a total of 43 infusions of VSTs for adenoviremia and/or adenoviral disease. Seven patients received donor-derived VSTs, 21 patients received third-party VSTs, and 2 received VSTs from both donor sources. Clinical responses were observed in 81% of patients, with a complete response in 58%. Epitope prediction and potential epitope identification for common HLA molecules helped elucidate HLA restriction in a subset of patients receiving third-party products. Intracellular interferon-γ expression in T cells in response to single peptides and response to cell lines stably transfected with a single HLA molecule demonstrated HLA-restricted CD4+ T-cell response, and these results correlated with clinical outcomes. Taken together, these data suggest that VSTs are a highly safe and effective therapy for the management of adenoviral infection in immunocompromised hosts. The trials were registered at www.clinicaltrials.gov as #NCT02048332 and #NCT02532452., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
39. Virus-specific T-cell therapy to treat BK polyomavirus infection in bone marrow and solid organ transplant recipients.
- Author
-
Nelson AS, Heyenbruch D, Rubinstein JD, Sabulski A, Jodele S, Thomas S, Lutzko C, Zhu X, Leemhuis T, Cancelas JA, Keller M, Bollard CM, Hanley PJ, Davies SM, and Grimley MS
- Subjects
- Bone Marrow, Cell- and Tissue-Based Therapy, Humans, Leukocytes, Mononuclear, BK Virus, Polyomavirus Infections therapy
- Abstract
BK polyomavirus (BKPyV) infection is a major complication of hematopoietic stem cell transplant (HSCT) and solid organ transplant (SOT). Treatment options are limited, poorly effective, and have significant toxicities. Cellular therapy using T cells directed against BKPyV is an emerging therapy, and we report efficacy in controlling BKPyV-associated disease in highly immunocompromised patients. Virus-specific T cells (VSTs) against BKPyV were manufactured using either blood from the patient's stem cell donor (donor-derived VSTs) or from unrelated donors (third-party VSTs). VSTs were used to treat BKPyV in 38 HSCT recipients and 3 SOT recipients between June 2017 and December 2019. Overall response rate was 86% in patients treated for BK viremia, 100% in patients treated for hemorrhagic cystitis, and 87% in patients treated for both BK viremia and hemorrhagic cystitis. No infusional toxicity, de novo graft-versus-host disease, or rejection of the organ occurred attributable to the VST infusion. BKPyV-specific immune responses were demonstrated by interferon-γ production by peripheral blood mononuclear cells postinfusion in response to BKPyV antigens. VSTs are a safe and potentially effective strategy to treat BKPyV and associated symptoms in recipients of HSCT and SOT. Cellular therapy should be considered for all patients with BKPyV and underlying immune suppression at risk of complications. This trial was registered at www.clinicaltrials.gov as #NCT02532452., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
40. Complement inhibition does not impair the clinical antiviral capabilities of virus-specific T-cell therapy.
- Author
-
Rubinstein JD, Zhu X, Lutzko C, Leemhuis T, Cancelas JA, Jodele S, Bollard CM, Hanley PJ, Davies SM, Grimley MS, and Nelson AS
- Subjects
- Cell- and Tissue-Based Therapy, Antiviral Agents pharmacology, CD8-Positive T-Lymphocytes
- Published
- 2020
- Full Text
- View/download PDF
41. EBV-directed viral-specific T-lymphocyte therapy for the treatment of EBV-driven lymphoma in two patients with primary immunodeficiency and DNA repair defects.
- Author
-
Rubinstein JD, Burns K, Absalon M, Lutzko C, Leemhuis T, Chandra S, Hanley PJ, Keller MD, Davies SM, Nelson A, and Grimley M
- Subjects
- Ataxia Telangiectasia etiology, Ataxia Telangiectasia pathology, Ataxia Telangiectasia Mutated Proteins genetics, Child, DNA Damage, Epstein-Barr Virus Infections virology, Female, Hodgkin Disease etiology, Hodgkin Disease pathology, Humans, Immunotherapy methods, Infant, Male, Mutation, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma etiology, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Prognosis, T-Lymphocytes immunology, Virus Activation, Ataxia Telangiectasia therapy, DNA Repair genetics, Epstein-Barr Virus Infections complications, Herpesvirus 4, Human genetics, Hodgkin Disease therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma therapy, T-Lymphocytes transplantation
- Abstract
Children with ataxia telangiectasia (AT), a primary immunodeficiency caused by mutations in ATM, which is critical for repairing DNA defects, are at risk for the development of hematologic malignancy, frequently driven by infection with Epstein-Barr virus (EBV). Conventional chemotherapy is poorly tolerated by patients with AT, with excessive toxicity even when doses are reduced. Here, we report on two patients with AT and EBV-positive neoplasms who were treated with EBV-targeted viral-specific T cells (VST). One patient had a prolonged complete response to VSTs while the other had a partial response. Therapy was well tolerated without infusion toxicity or graft-versus-host disease., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
42. CD38 bright CD8 + T Cells Associated with the Development of Acute GVHD Are Activated, Proliferating, and Cytotoxic Trafficking Cells.
- Author
-
Khandelwal P, Chaturvedi V, Owsley E, Lane A, Heyenbruch D, Lutzko CM, Leemhuis T, Grimley MS, Nelson AS, Davies SM, Jordan MB, and Marsh RA
- Subjects
- Adolescent, Adult, Allografts, CD8-Positive T-Lymphocytes pathology, Child, Child, Preschool, Cytomegalovirus immunology, Cytomegalovirus Infections immunology, Cytomegalovirus Infections pathology, Epstein-Barr Virus Infections immunology, Female, Graft vs Host Disease pathology, HLA-DR Antigens immunology, Herpesvirus 4, Human immunology, Humans, Infant, Male, ADP-ribosyl Cyclase 1 immunology, CD8-Positive T-Lymphocytes immunology, Graft vs Host Disease immunology, Hematopoietic Stem Cell Transplantation, Lymphocyte Activation, Membrane Glycoproteins immunology
- Abstract
We have previously reported that a peripheral blood absolute CD38
bright CD8+ effector memory T cell (TEM) population expansion of >35 cells/µL predicts the development of acute graft-versus-host disease (GVHD). We hypothesized that these T cells are activated, proliferating, and cytotoxic trafficking cells that are not a response to viral reactivation and may be involved in acute GVHD. We characterized peripheral blood T cell populations at the time of maximum CD38bright CD8+ TEM expansion in patients from our originally reported pediatric allogeneic hematopoietic cell transplantation recipient cohort. Samples were incubated with fluorochrome-conjugated antibodies directed against CD3, CD8, CD38, HLA-DR (T cell activation), Ki-67 (T cell proliferation), granzyme B (marker of cytotoxic T cells), CLA (skin trafficking), CCR5 (visceral trafficking), and CXCR6 (liver trafficking). We also incubated samples with Epstein-Barr virus (EBV) and cytomegalovirus (CMV) peptide pools and measured IFN-γ production by flow cytometry and performed EBV and CMV tetramer staining. Higher median proportions of cell expression of HLA-DR, Ki-67, granzyme B, CLA, CCR5, and CXCR6 were observed for CD38bright CD8+ T cells compared with CD38non bright T cells in patients with acute GVHD (P < .05) but not in patients without acute GVHD (P not significant). No IFN-γ production was observed after incubation with CMV and EBV peptide pools. EBV-specific tetramer populations of 6.85% and 3.17% were detected in 2 patients with acute GVHD, whereas a CMV-specific tetramer population of 3.77% was detected in 1 patient with acute GVHD. No EBV- or CMV-specific tetramer populations were detected in any patient without acute GVHD. We conclude that CD38+ T cells in patients with acute GVHD (P < .05) but not in patients without acute GVHD (P not significant). No IFN-γ production was observed after incubation with CMV and EBV peptide pools. EBV-specific tetramer populations of 6.85% and 3.17% were detected in 2 patients with acute GVHD, whereas a CMV-specific tetramer population of 3.77% was detected in 1 patient with acute GVHD. No EBV- or CMV-specific tetramer populations were detected in any patient without acute GVHD. We conclude that CD38bright T cells associated with the development of acute GVHD are activated, proliferating, and cytotoxic trafficking cells that do not appear to respond to CMV or EBV reactivation. Further studies are needed to determine whether these cells are directly involved in acute GVHD pathogenesis.+ T cells associated with the development of acute GVHD are activated, proliferating, and cytotoxic trafficking cells that do not appear to respond to CMV or EBV reactivation. Further studies are needed to determine whether these cells are directly involved in acute GVHD pathogenesis., (Copyright © 2019 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
43. Post-Transplant CD34 + Selected Stem Cell "Boost" for Mixed Chimerism after Reduced-Intensity Conditioning Hematopoietic Stem Cell Transplantation in Children and Young Adults with Primary Immune Deficiencies.
- Author
-
Chandra S, Bleesing JJ, Jordan MB, Grimley MS, Khandelwal P, Davies SM, Edwards S, Leemhuis T, and Marsh RA
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Female, Humans, Immunologic Deficiency Syndromes pathology, Infant, Infant, Newborn, Male, Young Adult, Antigens, CD34 therapeutic use, Chimerism chemically induced, Hematopoietic Stem Cell Transplantation methods, Immunologic Deficiency Syndromes therapy, Transplantation Conditioning methods
- Abstract
Mixed chimerism and eventual graft loss occurs in a proportion of children with primary immune deficiencies receiving alemtuzumab, fludarabine, and melphalan reduced-intensity conditioning (RIC) regimens before allogeneic hematopoietic stem cell transplantation (HSCT). We investigated the usefulness of a CD34
+ selected stem cell "boost" without conditioning to treat mixed chimerism in children and young adults who received predominantly an alemtuzumab, fludarabine, and melphalan RIC regimen for primary immune deficiencies and reported the outcomes. Patients with a primary immune deficiency disorder who were either enrolled on a prospective CD34+ boost study for treatment of mixed chimerism from 2011 to 2014 (n = 9) or treated with a CD34+ boost on a clinical basis from 2014 to 2016 (n = 3) were included in this analysis. Response to a CD34+ boost was defined as a rise in donor chimerism by ≥15% with donor chimerism of at least 20%, stabilization was defined as a rise in chimerism by <15% with donor chimerism ≥ 20%, and no response was defined as any decline in donor chimerism or need for a second HSCT after a CD34+ boost. Twelve patients received alemtuzumab, fludarabine, and melphalan. Median age was 4.5 years (range, .9 to 20.6), and median whole blood donor chimerism before the boost was 25% (range, 3% to 61%). Three patients (25%) met criteria for response, 1 patient (8%) was considered to have stabilization, and 8 patients (67%) had no response 12 months after the boost. None of the patients developed any complications from a CD34+ boost, including no acute graft-versus-host disease (GVHD). All patients are alive with a median follow-up of 32 months (range, 8 to 79). We conclude that a CD34+ selected stem cell boost can be considered for treatment of mixed chimerism after alemtuzumab, fludarabine, and melphalan RIC HSCT in children and young adults with primary immune deficiencies. Approximately one-third of patients can be expected to benefit from a CD34+ selected stem cell boost and may avoid the need for a second HSCT. Lack of any GVHD or toxicity makes a stem cell boost an attractive option compared with donor lymphocyte infusions for treatment of mixed chimerism., (Copyright © 2018 The American Society for Blood and Marrow Transplantation. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
44. Clinical methods of cryopreservation for donor lymphocyte infusions vary in their ability to preserve functional T-cell subpopulations.
- Author
-
Worsham DN, Reems JA, Szczepiorkowski ZM, McKenna DH, Leemhuis T, Mathew AJ, and Cancelas JA
- Subjects
- CD4-Positive T-Lymphocytes cytology, CD4-Positive T-Lymphocytes immunology, Cell Cycle physiology, Dimethyl Sulfoxide chemistry, Hematopoietic Stem Cell Transplantation, Humans, Immunophenotyping, T-Lymphocytes, Cytotoxic cytology, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Helper-Inducer cytology, T-Lymphocytes, Helper-Inducer immunology, T-Lymphocytes, Regulatory cytology, T-Lymphocytes, Regulatory immunology, Cryopreservation methods, Lymphocyte Transfusion methods
- Abstract
Background: Cryopreserved donor lymphocyte infusion (DLI) products are manufactured and administered to treat relapse after allogeneic hematopoietic stem cell transplantation. Reported clinical responses to DLIs vary broadly, even within the same group of patients. While there is an implicit recognition of the fact that different manufacturing protocols may have specific effects on different cell types, cryopreservation protocols are frequently derived from our experience in the cryopreservation of stem cell products and do not account for the heterogeneous functional nature of DLI T-cell populations. Here, we report the results of a prospective, multicenter trial on the effect of four different cryopreservation solutions that were used to freeze DLIs compared to control DLIs that were refrigerated overnight., Study Design and Methods: Cryopreserved postthawed and refrigerated specimens were analyzed side by side for their T-cell subpopulation content and viability, as well as T-cell proliferation, cytokine secretion, and cytotoxic activities., Results: This study indicates that "homemade" 10% dimethyl sulfoxide (DMSO) results in reduced viability of different CD4+ T-cell populations, including T-helper, T-cytotoxic, and T-regulatory populations, and a decrease in their proliferative and cytotoxic response to immunologically relevant stimuli, while the use of solutions containing 5% DMSO with intracellular-like cryoprotectant stabilizers maintains T-cell function at levels similar to refrigerated control samples., Conclusion: This study has important implications in determining the best cryoprotectant solution for specific clinical applications in allogeneic immunotherapy., (© 2017 AABB.)
- Published
- 2017
- Full Text
- View/download PDF
45. Radiation-free, alternative-donor HCT for Fanconi anemia patients: results from a prospective multi-institutional study.
- Author
-
Mehta PA, Davies SM, Leemhuis T, Myers K, Kernan NA, Prockop SE, Scaradavou A, O'Reilly RJ, Williams DA, Lehmann L, Guinan E, Margolis D, Baker KS, Lane A, and Boulad F
- Subjects
- Adolescent, Adult, Antilymphocyte Serum therapeutic use, Bone Marrow drug effects, Bone Marrow immunology, Bone Marrow pathology, Busulfan therapeutic use, Child, Child, Preschool, Cyclophosphamide therapeutic use, Fanconi Anemia immunology, Fanconi Anemia pathology, Female, Graft vs Host Disease immunology, Graft vs Host Disease pathology, Humans, Lymphocyte Depletion, Male, Myelodysplastic Syndromes immunology, Myelodysplastic Syndromes pathology, Prospective Studies, Siblings, Survival Analysis, T-Lymphocytes immunology, T-Lymphocytes pathology, Transplantation, Homologous, Treatment Outcome, Unrelated Donors, Vidarabine analogs & derivatives, Vidarabine therapeutic use, Fanconi Anemia therapy, Hematopoietic Stem Cell Transplantation, Myeloablative Agonists therapeutic use, Myelodysplastic Syndromes therapy, Transplantation Conditioning methods
- Abstract
Fanconi anemia (FA) is an inherited bone marrow failure syndrome characterized by chromosomal fragility, progressive marrow failure, and cancer predisposition. Hematopoietic cell transplantation (HCT) is curative for FA-related marrow failure or leukemia, but both radiation exposure during transplant and graft-versus-host disease (GVHD) may increase risk of later malignancies of the head and neck and anogenital area. In this study, we tested a radiation-free conditioning regimen with a T-cell-depleted graft to eliminate radiation exposure and minimize early and late toxicities of transplant. Forty-five patients (median age, 8.2 years; range 4.3-44) with FA underwent HCT between June 2009 and May 2014. The preparative regimen included busulfan, cyclophosphamide, fludarabine, and rabbit anti-thymocyte globulin. Busulfan levels were monitored to avoid excess toxicity. All grafts were CD34-selected/T-cell-depleted using the CliniMacs CD34 columns (Miltenyi). Thirty-four patients (75.6%) with marrow failure and 11 (24.4%) with myelodysplastic syndrome underwent HCT using matched unrelated (n = 25, 55.5%), mismatched unrelated (n = 14, 31.1%), or mismatched related donors (n = 6, 13.4%). One year probabilities of overall and disease-free survival for the entire cohort, including patients with myeloid malignancy and those receiving mismatched related/haploidentical grafts, were 80% (±6%) and 77.7% (±6.2%), respectively (median follow-up 41 months). All young children (<10 years of age) undergoing HCT for marrow failure using low-dose busulfan-containing regimen survived. No patients developed acute grade 3-4 GVHD. Sequential reduction of busulfan dose was successfully achieved per study design. Our results show excellent outcomes in patients with FA undergoing alternative donor HCT without radiation exposure. The study is registered to www.clinicaltrials.gov as #NCT01082133., (© 2017 by The American Society of Hematology.)
- Published
- 2017
- Full Text
- View/download PDF
46. Experience with Alemtuzumab, Fludarabine, and Melphalan Reduced-Intensity Conditioning Hematopoietic Cell Transplantation in Patients with Nonmalignant Diseases Reveals Good Outcomes and That the Risk of Mixed Chimerism Depends on Underlying Disease, Stem Cell Source, and Alemtuzumab Regimen.
- Author
-
Marsh RA, Rao MB, Gefen A, Bellman D, Mehta PA, Khandelwal P, Chandra S, Jodele S, Myers KC, Grimley M, Dandoy C, El-Bietar J, Kumar AR, Leemhuis T, Zhang K, Bleesing JJ, Jordan MB, Filipovich AH, and Davies SM
- Subjects
- Adolescent, Adult, Alemtuzumab, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Child, Child, Preschool, Chimerism, Female, Hematopoietic Stem Cell Transplantation methods, Humans, Male, Melphalan administration & dosage, Transplantation Conditioning methods, Treatment Outcome, Vidarabine administration & dosage, Vidarabine therapeutic use, Young Adult, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Melphalan therapeutic use, Vidarabine analogs & derivatives
- Abstract
Alemtuzumab, fludarabine, and melphalan reduced-intensity conditioning (RIC) regimens are increasingly used for the hematopoietic cell transplantation (HCT) of pediatric and young adult patients with nonmalignant diseases. Early experience suggests that these regimens are associated with good survival but a high incidence of mixed chimerism, which we have previously shown to be influenced by the alemtuzumab schedule. We hypothesized that the underlying diagnosis and donor graft source would also affect the development of mixed chimerism and that the majority of patients would survive RIC HCT without graft loss. To examine this, we conducted a retrospective study of 206 patients with metabolic diseases, non-Fanconi anemia marrow failure disorders, and primary immune deficiencies who underwent 210 consecutive RIC HCT procedures at Cincinnati Children's Hospital. Ninety-seven percent of the patients engrafted. Mixed donor and recipient chimerism developed in 46% of patients. Patients with marrow failure had a low risk of mixed chimerism (hazard ratio [HR], .208; 95% confidence interval [CI], .061 to .709; P = .012). The risk of mixed chimerism was high in patients who received a cord blood graft (HR, 3.122; 95% CI, 1.236 to 7.888; P = .016). As expected, patients who received a proximal or higher dose per kilogram of alemtuzumab schedule also experienced higher rates of mixed chimerism (all HR > 2, all P < .05). At the time of last follow-up (median, 654 days; range, 13 to 3337), over 75% of patients had greater than 90% whole blood donor chimerism. A second transplantation was performed in 5% of patients. Three-year survival without retransplantation was 84% (95% CI, 71% to 98%) for patients who underwent transplantation with an HLA-matched sibling donor. Survival without retransplantation was negatively affected by lack of a matched related donor, increasing age, and development of grades III and IV acute graft-versus-host disease. We conclude that alemtuzumab, fludarabine, and melphalan RIC HCT offers good results for many patients and that the risk of developing mixed chimerism is influenced by underlying diagnosis, graft source, and alemtuzumab dosing., (Copyright © 2015 American Society for Blood and Marrow Transplantation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
47. An intermediate alemtuzumab schedule reduces the incidence of mixed chimerism following reduced-intensity conditioning hematopoietic cell transplantation for hemophagocytic lymphohistiocytosis.
- Author
-
Marsh RA, Kim MO, Liu C, Bellman D, Hart L, Grimley M, Kumar A, Jodele S, Myers KC, Chandra S, Leemhuis T, Mehta PA, Bleesing JJ, Davies SM, Jordan MB, and Filipovich AH
- Subjects
- Adolescent, Adult, Alemtuzumab, Child, Child, Preschool, Female, Hematopoietic Stem Cell Transplantation adverse effects, Humans, Incidence, Infant, Infant, Newborn, Lymphohistiocytosis, Hemophagocytic drug therapy, Lymphohistiocytosis, Hemophagocytic surgery, Male, Transplantation Chimera, Transplantation Conditioning adverse effects, Transplantation, Homologous, Young Adult, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Agents administration & dosage, Hematopoietic Stem Cell Transplantation methods, Lymphohistiocytosis, Hemophagocytic therapy, Transplantation Conditioning methods
- Abstract
Reduced-intensity conditioning (RIC) improves the outcomes of hematopoietic cell transplantation (HCT) in patients with hemophagocytic lymphohistiocytosis (HLH). Proximal (ie, close to graft infusion) dosing of alemtuzumab is associated with a high incidence of mixed chimerism, whereas distal (ie, distant from graft infusion) dosing is associated with less mixed chimerism but more acute graft-versus-host disease (GVHD). The alemtuzumab dose per kilogram of body weight also influences these outcomes. We hypothesized that an intermediate alemtuzumab dosing schedule would reduce mixed chimerism and maintain a low incidence of acute GVHD. In this study, 24 consecutive HCTs were performed in patients with HLH or a related disorder using a novel intermediate alemtuzumab schedule of 1 mg/kg starting on day -14. The cumulative incidences (CIs) of mixed chimerism, upfront acute GVHD grades II-IV, and receipt of additional hematopoietic cell products after HCT were compared in patients treated with a distal alemtuzumab schedule (n = 15) and those treated with a proximal alemtuzumab schedule (n = 33). All patients received fludarabine and melphalan. The CI of mixed chimerism was 31% in the intermediate group, 72% in the proximal group (P < .01), and 75% in the distal group patients who received ≥2 mg/kg alemtuzumab (P = .03). The CI of acute GVHD grades II-IV before the development of mixed chimerism was 4% in the intermediate group, 0% in the proximal group, and 13% in the distal group (P = .04, proximal versus distal). The 1-year CI of administration of additional hematopoietic cell products for mixed chimerism (donor lymphocyte infusion ± hematopoietic stem cell boost ± repeat HCT) was 14% in the intermediate group, 53% in the proximal group (P = .01), and 38% in the distal ≥2 mg/kg alemtuzumab group (P = .02). Our findings indicate that intermediate RIC reduces the incidence of mixed chimerism, is associated with a low incidence of upfront acute GVHD, and decreases the need for additional hematopoietic cell products after HCT., (Copyright © 2013 American Society for Blood and Marrow Transplantation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
48. Stem cell collection and gene transfer in Fanconi anemia.
- Author
-
Kelly PF, Radtke S, von Kalle C, Balcik B, Bohn K, Mueller R, Schuesler T, Haren M, Reeves L, Cancelas JA, Leemhuis T, Harris R, Auerbach AD, Smith FO, Davies SM, and Williams DA
- Subjects
- Adolescent, Antigens, CD34 metabolism, Bone Marrow metabolism, Child, Child, Preschool, DNA, Complementary genetics, Fanconi Anemia metabolism, Fanconi Anemia pathology, Fanconi Anemia therapy, Fanconi Anemia Complementation Group A Protein genetics, Humans, Infant, Cell Separation methods, Fanconi Anemia genetics, Genetic Therapy adverse effects, Stem Cells cytology, Transgenes genetics
- Abstract
Fanconi anemia (FA) is a rare genetic syndrome characterized by progressive bone marrow failure (BMF), congenital anomalies, and a predisposition to malignancy. Successful gene transfer into hematopoietic stem cells (HSCs) could reverse BMF in this disease. We developed clinical trials to determine whether a sufficient number of CD34(+) stem cells could be collected for gene modification and to evaluate the safety and efficacy of HSC-corrective gene transfer in FA genotype A (FANCA) patients. Here, we report that FA patients have significant depletion of their BM CD34(+) cell compartment even before severe pancytopenia is present. However, oncoretroviral-mediated ex vivo gene transfer was efficient in clinical scale in FA-A cells, leading to reversal of the cellular phenotype in a significant percentage of CD34(+) cells. Re-infusion of gene-corrected products in two patients was safe and well tolerated and accompanied by transient improvements in hemoglobin and platelet counts. Gene correction was transient, likely owing to the low dose of gene-corrected cells infused. Our early experience shows that stem cell collection is well tolerated in FA patients and suggests that collection be considered as early as possible in patients who are potential candidates for future gene transfer trials.
- Published
- 2007
- Full Text
- View/download PDF
49. A phase I trial of autologous cytokine-induced killer cells for the treatment of relapsed Hodgkin disease and non-Hodgkin lymphoma.
- Author
-
Leemhuis T, Wells S, Scheffold C, Edinger M, and Negrin RS
- Subjects
- Adult, Aged, Antigens, CD analysis, Cell Culture Techniques, Cells, Cultured, Cytotoxicity, Immunologic, Female, Hodgkin Disease complications, Humans, Immunotherapy, Adoptive adverse effects, Killer Cells, Lymphokine-Activated cytology, Lymphocyte Transfusion adverse effects, Lymphocyte Transfusion methods, Lymphoma, Non-Hodgkin complications, Male, Middle Aged, Salvage Therapy methods, Transplantation, Autologous, Treatment Outcome, Hodgkin Disease therapy, Immunotherapy, Adoptive methods, Killer Cells, Lymphokine-Activated transplantation, Lymphoma, Non-Hodgkin therapy
- Abstract
We have previously reported on the ex vivo generation of cytotoxic effector cells, termed cytokine-induced killer (CIK) cells, that have both in vitro and in vivo antitumor activity in murine models. We now report on our efforts for the large-scale expansion of CIK cells and also present preliminary results from a phase I clinical trial. Nine patients with advanced Hodgkin disease (n = 7) and non-Hodgkin lymphoma (n = 2), all of whom had relapsed after an autologous transplantation, were treated with escalating doses of CIK cells (3 patients at each dose level of 1 x 10(9) , 5 x 10(9) , or 1 x 10(10) cells). The CIK cells were produced by culturing unselected cells from steady-state apheresis products with interferon gamma, OKT3, and interleukin 2. After 21 days in culture, with the addition of fresh media and interleukin 2 every 3 to 4 days, the median culture was 97% viable (range, 61%-100%), 98% CD3 + (range, 66%-99%), 76% CD8 + (range, 27%-96%), 23% CD4 + (range, 6%-78%), 20% CD3 + CD56 + (range, 8%-58%), and <1% CD16 + 56 + (range, 0.2%-7.7%). The CD3 + CD56 + cells have previously been shown to exhibit the most cytotoxic activity. The absolute number of CD3 + CD56 + cells typically expanded 290-fold (range, 3- to 4000-fold) under these culture conditions. In vitro cytotoxic activity was measured against a human B-cell tumor line (OCI-Ly8). At a 40:1 effector-target cell ratio, CIK cells killed 32% (range, 2%-69%) of the target cells. A total of 21 infusions were administered to 9 patients. The number of CIK cells infused ranged from 1.0 x 10(9) to 1.0 x 10(10) per treatment. Toxicity was minimal, and there were no immediate adverse reactions to the infusions. Two patients had partial responses, and 2 patients had stabilization of disease: 1 for more than 18 months. Considering that these were heavily pretreated patients with advanced hematologic malignancies, we believe that CIK cells expanded in this fashion may have utility for the treatment of high-risk patients with evidence of minimal residual disease after autologous transplantation.
- Published
- 2005
- Full Text
- View/download PDF
50. Low risk of graft-versus-host disease with transplantation of CD34 selected peripheral blood progenitor cells from alternative donors for Fanconi anemia.
- Author
-
Boyer MW, Gross TG, Loechelt B, Leemhuis T, Filipovich A, and Harris RE
- Subjects
- Adolescent, Child, Child, Preschool, Female, Humans, Infant, Lymphocyte Depletion, Male, Risk, Tissue Donors, Transplantation Conditioning, Antigens, CD34 analysis, Fanconi Anemia therapy, Graft vs Host Disease prevention & control, Peripheral Blood Stem Cell Transplantation adverse effects, Peripheral Blood Stem Cell Transplantation mortality
- Abstract
Objectives: Transplant results for Fanconi anemia with alternative-donor bone marrow transplantation currently entail a high incidence of graft failure and graft-versus-host disease (GVHD). The authors sought to improve outcome in this disease category with alternative donors with a 5-6/6 antigen match by transplantation of highly purified peripheral blood progenitor cells (PBPC) using the Isolex 300i v2.5 device as a means of T-cell depletion to lessen the risk of GVHD., Methods: All Fanconi anemia patients (n = 8) received the same preparative regimen that included total body irradiation (450 cGy), Cytoxan (20 mg/kg), ATGAM, and fludarabine (120 mg/m2). The cell dose of CD34+ cells was a median of 11.4 x 10(6)/kg; the cell dose of CD3+ cells was a median of 1.9 x 10(4)/kg. Primary engraftment was rapid in all patients, with neutrophil recovery occurring at a median of day 10 and platelet count more than 50,000 on day 27. Two patients subsequently had secondary graft failure. Despite lack of cyclosporine GVHD prophylaxis, only two patients developed acute GVHD (both grade I), and no patients developed chronic GVHD. Three patients died: one at day 59 secondary to disseminated fungal infection, the second at day 196 during a second transplant, and the third at day 202 due to graft failure. With a median follow-up of 12 months, the overall survival was 58 +/- 18%., Conclusions: Transplantation of CD34-selected PBPCs from alternative donors results in a very low risk of GVHD in patients with Fanconi anemia.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.