Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Pract Radiat Oncol ; 12(6): e476-e480, 2022.
Article in English | MEDLINE | ID: mdl-35598860

ABSTRACT

We present the case of a 56-year-old female with a diagnosis of acute T-cell lymphoblastic leukemia who received myeloablative conditioning for bone marrow transplant with total body irradiation (TBI) using volumetric modulated arc therapy (VMAT) to the upper body and anterior-posterior/posterior-anterior (AP/PA) open fields to the lower body followed by hematopoietic stem cell transplant. Her clinical course was complicated by high-grade pulmonary toxic effects 55 days after treatment that resulted in death. We discuss the case, planning considerations by radiation oncologists and radiation physicists, and the multidisciplinary medical management of this patient.


Subject(s)
Radiotherapy, Intensity-Modulated , Whole-Body Irradiation , Humans , Female , Middle Aged , Whole-Body Irradiation/adverse effects , Whole-Body Irradiation/methods , Radiotherapy, Intensity-Modulated/adverse effects , Radiotherapy, Intensity-Modulated/methods , Transplantation Conditioning/adverse effects , Transplantation Conditioning/methods , Vidarabine/adverse effects , Retrospective Studies
3.
Front Immunol ; 11: 636, 2020.
Article in English | MEDLINE | ID: mdl-32373119

ABSTRACT

Following allogeneic blood and marrow transplantation (BMT), graft-versus-host disease (GvHD) continues to represent a significant cause of treatment failure, despite the routine use of conventional, mainly calcineurin inhibitor-based prophylaxis. Recently, post-transplant cyclophosphamide (PTCy) has emerged as a safe and efficacious alternative. First, omitting the need for ex vivo T-cell depletion in the setting of haploidentical transplantation, growing evidence supports PTCy role in GvHD prevention in matched-related and matched-unrelated transplants. Through improved understanding of GvHD pathophysiology and advancements in drug development, PTCy emerges as a unique opportunity to design calcineurin inhibitor-free strategies by integrating agents that target different stages of GvHD development.


Subject(s)
Bone Marrow Transplantation , Cyclophosphamide/therapeutic use , Graft vs Host Disease/prevention & control , Immunosuppressive Agents/therapeutic use , Postoperative Complications/prevention & control , Animals , Graft vs Host Disease/etiology , HLA Antigens/genetics , HLA Antigens/metabolism , Histocompatibility , Histocompatibility Testing , Humans , Transplantation, Haploidentical , Unrelated Donors
4.
Exp Hematol ; 48: 50-57, 2017 04.
Article in English | MEDLINE | ID: mdl-28007655

ABSTRACT

There is an abiding need for innovative approaches to the prevention of graft-versus-host disease (GvHD) following allogeneic hematopoietic stem cell transplantation (HSCT). Interest in prevention of GvHD by dendritic cell (DC) suppression has re-emerged since the introduction of proteasome inhibitors into clinical practice. Ixazomib is an orally bioavailable proteasome inhibitor with a rapid proteasome dissociation rate. We studied the effects of ixazomib on human DC maturation, viability, and cytokine production in vitro. We also determined the effects of ixazomib in a murine GvHD model. Although ixazomib suppressed naïve human DC maturation, it had only a limited effect on cell viability. Ixazomib decreased pro-inflammatory cytokine production of resting DCs. This effect was diminished or reversed when DCs were pre-stimulated. In vivo, ixazomib administered post-transplantation on days +1 and +4 or days -1, +2, and +5 ameliorated GvHD in comparison to the GvHD group. Although a fraction of mice treated according to the prolonged schedule died abruptly after the day +5 treatment, both schedules resulted in improved overall survival. When we examined the effects of ixazomib on splenic cells and serum cytokines, we found that ixazomib exerted complex schedule-dependent immunomodulatory effects. Our study provides a rationale for the potential use of ixazomib in the prevention of GvHD.


Subject(s)
Boron Compounds/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Glycine/analogs & derivatives , Graft vs Host Disease/etiology , Protease Inhibitors/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Glycine/pharmacology , Graft vs Host Disease/drug therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunomodulation/drug effects , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
5.
Biol Blood Marrow Transplant ; 23(2): 255-261, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27888016

ABSTRACT

Lapses in the prevention of graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (HSCT) warrant novel approaches. Such approaches include, among others, the use of post-transplantation cyclophosphamide (PTC) and proteasome inhibitors. Although PTC alone consistently produces low rates of chronic GVHD, the incidence of acute GVHD remains significant. Inversely, prolonged post-transplantation administration of proteasome inhibitors carries a risk of paradoxical aggravation of GVHD. We examined whether the combination of cyclophosphamide and ixazomib addresses the limitations of each of these agents when used alone to prevent GVHD in mice subjected to allogeneic HSCT across MHC barriers. We chose ixazomib, an orally bioavailable proteasome inhibitor, because of its favorable physiochemical characteristics. The combination of cyclophosphamide and ixazomib improved overall survival of mice in comparison to an untreated control group and to groups receiving either cyclophosphamide alone or ixazomib alone. Furthermore, cyclophosphamide prevented the surge of IL-1ß, GVHD aggravation, and sudden death associated with prolonged administration of ixazomib after HSCT. Finally, we demonstrated that although ixazomib was administered before cyclophosphamide, it did not impair the preferential depletion of proliferating as opposed to resting donor T cells. Our data suggest that the combination of cyclophosphamide and ixazomib for the prevention of GVHD after allogeneic HSCT is promising and merits further investigation in clinical trials.


Subject(s)
Boron Compounds/therapeutic use , Cyclophosphamide/therapeutic use , Glycine/analogs & derivatives , Graft vs Host Disease/prevention & control , Proteasome Inhibitors/therapeutic use , Animals , Bone Marrow Transplantation/adverse effects , Boron Compounds/administration & dosage , Boron Compounds/pharmacology , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , Cytokines/blood , Drug Administration Schedule , Drug Evaluation, Preclinical , Drug Synergism , Drug Therapy, Combination , Female , Glycine/administration & dosage , Glycine/pharmacology , Glycine/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Host Disease/pathology , Intestine, Small/pathology , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Proteasome Inhibitors/administration & dosage , Proteasome Inhibitors/pharmacology , Radiation Chimera , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/transplantation
6.
Exp Hematol ; 44(9): 771-777, 2016 09.
Article in English | MEDLINE | ID: mdl-27224851

ABSTRACT

Allogeneic hematopoietic stem cell transplantation is the standard treatment for a variety of benign and malignant conditions. However, graft-versus-host disease (GvHD) continues to present a major barrier to the success and wide applicability of this procedure. Although current GvHD prevention and treatment regimens exclusively target T cells, bortezomib, a reversible proteasome inhibitor, possesses unique immune regulatory activities that span a wide variety of cellular processes of T and dendritic cells essential for the development of GvHD. Herein, we review the current understanding of the effects of bortezomib in vitro and in animal models and summarize the clinical data relevant to its use in the prevention and treatment of GvHD. We conclude with an outline of the remaining challenges and opportunities to optimize bortezomib's potential role in this setting.


Subject(s)
Bortezomib/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Animals , Apoptosis/drug effects , Bortezomib/pharmacology , Clinical Trials as Topic , Cytokines/biosynthesis , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , Humans , Lymphocyte Activation/drug effects , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transplantation, Homologous , Treatment Outcome
7.
Biol Blood Marrow Transplant ; 21(7): 1315-20, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25765556

ABSTRACT

An effective graft-versus-host disease (GVHD) preventative approach that preserves the graft-versus-tumor effect after allogeneic hematopoietic stem cell transplantation (HSCT) remains elusive. Standard GVHD prophylactic regimens suppress T cells indiscriminately and are suboptimal. Conversely, post-transplantation high-dose cyclophosphamide selectively destroys proliferating alloreactive T cells, allows the expansion of regulatory T cells, and induces long-lasting clonal deletion of intrathymic antihost T cells. It has been successfully used to prevent GVHD after allogeneic HSCT. Bortezomib has antitumor activity on a variety of hematological malignancies and exhibits a number of favorable immunomodulatory effects that include inhibition of dendritic cells. Therefore, an approach that combines post-transplantation cyclophosphamide and bortezomib seems attractive. Herein, we report the results of a phase I study examining the feasibility and safety of high-dose post-transplantation cyclophosphamide in combination with bortezomib in patients undergoing allogeneic peripheral blood HSCT from matched siblings or unrelated donors after reduced-intensity conditioning. Cyclophosphamide was given at a fixed dose (50 mg/kg on days +3 and +4). Bortezomib dose was started at .7 mg/m2, escalated up to 1.3 mg/m2, and was administered on days 0 and +3. Patients receiving grafts from unrelated donors also received rabbit antithymocyte globulin. The combination was well tolerated and allowed prompt engraftment in all patients. The incidences of acute GVHD grades II to IV and grades III and IV were 20% and 6.7%, respectively. With a median follow-up of 9.1 months (range, 4.3 to 26.7), treatment-related mortality was 13.5% with predicted 2-year disease-free survival and overall survival of 55.7% and 68%, respectively. The study suggests that the combination of post-transplantation cyclophosphamide and bortezomib is feasible and may offer an effective and practical GVHD prophylactic regimen. The combination, therefore, merits further examination.


Subject(s)
Bortezomib/therapeutic use , Cyclophosphamide/therapeutic use , Graft vs Host Disease/prevention & control , Hematologic Neoplasms/therapy , Peripheral Blood Stem Cell Transplantation/methods , Transplantation Conditioning , Acute Disease , Adult , Aged , Antilymphocyte Serum/therapeutic use , Drug Administration Schedule , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Hematologic Neoplasms/immunology , Hematologic Neoplasms/mortality , Hematologic Neoplasms/pathology , Humans , Male , Middle Aged , Myeloablative Agonists/therapeutic use , Peripheral Blood Stem Cell Transplantation/adverse effects , Risk , Siblings , Survival Analysis , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transplantation, Homologous , Treatment Outcome , Unrelated Donors
8.
Biol Blood Marrow Transplant ; 21(4): 604-11, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25240817

ABSTRACT

Cyclophosphamide's lack of hematopoietic stem cell toxicity and its unique effects on the immune system have prompted several investigators to explore its potential for the prevention of graft-versus-host disease (GVHD). In haploidentical hematopoietic stem cell transplants, post-transplant cyclophosphamide together with standard prophylaxis reduces the incidence of GVHD to acceptable rates without the need for T cell depletion. In matched related and unrelated donor settings, cyclophosphamide alone has produced encouraging results. In particular, the low incidence of chronic GVHD is noteworthy. Here, we present a review of the current understanding of the mechanism of action of post-transplant cyclophosphamide and summarize the clinical data on its use for the prevention of GVHD.


Subject(s)
Cyclophosphamide/therapeutic use , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/therapeutic use , Lymphocyte Depletion , Unrelated Donors , Allografts , Graft vs Host Disease/etiology , Humans
9.
Transpl Immunol ; 30(1): 40-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24262132

ABSTRACT

Impairing dendritic cell (DC) function to prevent graft versus host disease (GvHD) is an appealing concept. DC antigen presentation is NF-κB pathway-dependent and bortezomib might therefore play a role in preventing alloreactivity. We obtained DC from the blood of patients enrolled in a phase I study using post-transplant cyclophosphamide and bortezomib for prevention of GvHD. Control samples were obtained from patients receiving standard GvHD prevention regimen. Pre-treatment samples were also collected from enrolled patients. DC isolated on days +1, +4, and +7 showed progressive decrease in the expression of maturation markers in comparison to control. In a DC-CD4+ mixed lymphocyte reaction (MLR) where DC isolated from the recipient blood before graft infusion were the stimulator cells, T cell proliferation measured by bromodeoxyuridine (BrdU) integration was decreased in samples obtained on days +14 and +21 in comparison to control group. Finally, measured by real-time PCR, the expression of IκB progressively increased while the expression of NF-κB decreased in DC on days +1, +4, and +7, in comparison to pre-treatment paired controls. We conclude that our data further justify exploring the role of bortezomib in GvHD prevention and propose a novel mechanism of action of bortezomib in DC.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Graft vs Host Disease/prevention & control , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Antigen Presentation/drug effects , Antigen Presentation/immunology , Boronic Acids/administration & dosage , Bortezomib , Cell Differentiation/drug effects , Cell Proliferation , Cells, Cultured , Cyclophosphamide/administration & dosage , Gene Expression Regulation/drug effects , Graft vs Host Disease/immunology , Humans , I-kappa B Proteins/genetics , Immunosuppressive Agents/administration & dosage , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , NF-kappa B/genetics , Postoperative Care , Pyrazines/administration & dosage
10.
Transpl Immunol ; 29(1-4): 1-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24103732

ABSTRACT

Dendritic cells (DC) play a central role in the pathophysiology of graft versus host disease (GvHD). Their antigen presenting capacity is nuclear factor κB- (NF-κB) dependent. Consequently, DC have emerged as a potential target for the prevention of GvHD and clinical trials with bortezomib are underway. We explored the activity of novel proteasome and immunoproteasome inhibitors on healthy volunteer peripheral blood DC. After incubation with the drug or drug combination, DC were stimulated with lipopolysaccharide, stained for maturation surface markers and then analyzed by flow cytometry. We found that the different molecule(s) inhibited DC maturation marker expression to variable degrees, with the constitutive proteasome-selective agent being the least active. In a DC and allogeneic CD4+ mixed lymphocyte reaction, DC incubation with the studied proteasome and immunoproteasome inhibitor(s), impeded T cell proliferation as measured by BrDU incorporation. Finally, we found that DC incubation with the drug(s) enhanced IκB expression and that oprozomib inhibited NF-κB expression. We concluded that based on its activity and oral bioavailability, oprozomib merits further investigation in an animal GvHD prevention model. We also suggest that altering IκB and NF-κB expressions may, in DC, represent a new mechanism of action of proteasome and immunoproteasome inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Dendritic Cells/metabolism , Gene Expression Regulation/drug effects , I-kappa B Kinase/biosynthesis , NF-kappa B/biosynthesis , Proteasome Inhibitors/pharmacology , Pyrazines/pharmacology , Bortezomib , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Dendritic Cells/pathology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...