Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Ann Clin Transl Neurol ; 11(2): 450-465, 2024 02.
Article in English | MEDLINE | ID: mdl-38204286

ABSTRACT

OBJECTIVE: Repeated intravenous administration of anti-CD20 depleting monoclonal antibodies 6 months apart is among the highly effective treatment options in multiple sclerosis (MS). Here, we aimed to investigate peripheral immune cell subset depletion kinetics following either rituximab (RTX) or ocrelizumab (OCR) infusions in people with MS (pwMS). METHODS: We studied pwMS treated de-novo with either RTX (n = 7) or OCR (n = 8). The examinations were scheduled before the initiation of anti-CD20 therapy and every 12 weeks for up to 15 months. Immunophenotyping of immune cell subsets in peripheral blood was performed by multiparametric fluorescence cytometry. RESULTS: A significant, persistent decrease of CD19+ B cells was observed already with the first anti-CD20 infusion (p < 0.0001). A significant proportional reduction of memory B cells within the B-cell pool was achieved only after two treatment cycles (p = 0.005). We observed a proportional increase of immature (p = 0.04) and naive B cells (p = 0.004), again only after the second treatment cycle. As for the peripheral T-cell pool, we observed a continuous proportional increase of memory T helper (TH) cells/central memory TH cells (p = 0.02/p = 0.008), while the number of regulatory T cells (Treg) decreased (p = 0.007). The percentage of B-cell dependent TH17.1 central memory cells dropped after the second treatment cycle (p = 0.02). No significant differences in the depletion kinetics between RTX and OCR were found. INTERPRETATION: Peripheral immune cell profiling revealed more differentiated insights into the prompt and delayed immunological effects of repeated intravenous anti-CD20 treatment. The observation of proportional changes of some pathogenetically relevant immune cell subsets only after two infusion cycles deserves further attention.


Subject(s)
Multiple Sclerosis , Humans , Rituximab/pharmacology , Rituximab/therapeutic use , Antibodies, Monoclonal/pharmacology , B-Lymphocytes , Antigens, CD19/metabolism , Antigens, CD19/pharmacology
2.
Ann Pharmacother ; 55(3): 390-405, 2021 03.
Article in English | MEDLINE | ID: mdl-32698673

ABSTRACT

OBJECTIVE: To evaluate the literature for axicabtagene ciloleucel (axi-cel), a first-in-class chimeric antigen receptor (CAR) T-cell therapy, in the treatment of relapsed/refractory (r/r) large B-cell lymphoma (LBCL). DATA SOURCES: We conducted a PubMed (inception to June 22, 2020) and ClinicalTrials.gov search using the following terms: CD19, chimeric antigen receptor, and lymphoma. STUDY SELECTION AND DATA EXTRACTION: All retrospective and prospective studies evaluating the use of axi-cel in LBCL were reviewed. DATA SYNTHESIS: In the pivotal ZUMA-1 trial, axi-cel exhibited unprecedented overall and complete response rates of 83% and 58%, respectively. With a median follow-up of 27.1 months, 39% of patients had ongoing responses. Furthermore, postmarketing retrospective analyses found similar response rates in a more clinically diverse LBCL patient population. Novel CAR T-cell therapy elicits unique and potentially life-threatening toxicities that include cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Studies reported grade ≥3 CRS in 7% to 14% of patients and grade ≥3 ICANS in 31% to 55% of patients. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE: Axi-cel was the first US Food and Drug Administration-approved genetically engineered autologous CAR T-cell agent in r/r LBCL, representing an important milestone and paradigm shift in cancer treatment. Adoptive T-cell immunotherapy is a breakthrough treatment modality requiring careful patient selection, multidisciplinary collaboration, comprehensive patient counseling, and expert training to ensure optimal treatment. CONCLUSIONS: The initial and ongoing results with axi-cel are encouraging, but long-term safety and efficacy data are lacking. Additional studies are required to identify axi-cel's ideal place in LBCL therapy.


Subject(s)
Antigens, CD19/therapeutic use , Immunotherapy, Adoptive/methods , Lymphoma, Large B-Cell, Diffuse/drug therapy , Receptors, Chimeric Antigen/metabolism , Antigens, CD19/pharmacology , Biological Products , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Prospective Studies , Recurrence , Retrospective Studies
3.
JCI Insight ; 5(14)2020 07 23.
Article in English | MEDLINE | ID: mdl-32525846

ABSTRACT

Regulatory T cells (Tregs) play essential roles in maintaining immunological self-tolerance and preventing autoimmunity. The adoptive transfer of antigen-specific Tregs has been expected to be a potent therapeutic method for autoimmune diseases, severe allergy, and rejection in organ transplantation. However, effective Treg therapy has not yet been established because of the difficulty in preparing a limited number of antigen-specific Tregs. Chimeric antigen receptor (CAR) T cells have been shown to be a powerful therapeutic method for treating B cell lymphomas, but application of CAR to Treg-mediated therapy has not yet been established. Here, we generated CD19-targeted CAR (CD19-CAR) Tregs from human PBMCs (hPBMCs) and optimized the fraction of the Treg source as CD4+CD25+CD127loCD45RA+CD45RO-. CD19-CAR Tregs could be expanded in vitro while maintaining Treg properties, including high expression of the latent form of TGF-ß. CD19-CAR Tregs suppressed IgG antibody production and differentiation of B cells via a TGF-ß-dependent mechanism. Unlike conventional CD19-CAR CD8+ T cells, CD19-CAR Tregs suppressed antibody production in immunodeficient mice that were reconstituted with hPBMCs, reducing the risk of graft-versus-host disease. Therefore, the adoptive transfer of CD19-CAR Tregs may provide a novel therapeutic method for treating autoantibody-mediated autoimmune diseases.


Subject(s)
Antigens, CD19/immunology , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer/methods , Animals , Antigens, CD19/adverse effects , Antigens, CD19/genetics , Antigens, CD19/pharmacology , Autoimmunity/genetics , Autoimmunity/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Epitopes , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Humans , Immune Tolerance/genetics , Immunoglobulin G/immunology , Lymphoma, B-Cell/complications , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Mice , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/therapeutic use , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/genetics
4.
Cells ; 9(5)2020 05 15.
Article in English | MEDLINE | ID: mdl-32429189

ABSTRACT

BACKGROUND: T lymphocyte collection through leukapheresis is an essential step for chimeric antigen receptor T (CAR-T) cell therapy. Timing of apheresis is challenging in heavily pretreated patients who suffer from rapid progressive disease and receive T cell impairing medication. METHODS: A total of 75 unstimulated leukaphereses were analyzed including 45 aphereses in patients and 30 in healthy donors. Thereof, 41 adult patients with Non-Hodgkin's lymphoma (85%) or acute lymphoblastic leukemia (15%) underwent leukapheresis for CAR-T cell production. RESULTS: Sufficient lymphocytes were harvested from all patients even from those with low peripheral lymphocyte counts of 0.18/nL. Only four patients required a second leukapheresis session. Leukapheresis products contained a median of 98 × 108 (9 - 341 × 108) total nucleated cells (TNC) with 38 × 108 (4 - 232 × 108) CD3+ T cells. Leukapheresis products from healthy donors as well as from patients in complete remission were characterized by high TNC and CD3+ T lymphocyte counts. CAR-T cell products could be manufactured for all but one patient. CONCLUSIONS: Sufficient yield of lymphocytes for CAR-T cell production is feasible also for patients with low peripheral blood counts. Up to 12-15 L blood volume should be processed in patients with absolute lymphocyte counts ≤ 1.0/nL.


Subject(s)
Leukapheresis , Lymphoma, Non-Hodgkin/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/cytology , Adult , Aged , Antigens, CD19/pharmacology , Antigens, CD19/therapeutic use , Biological Products , CD3 Complex/metabolism , Female , Humans , Immunotherapy, Adoptive , Lymphocyte Count , Lymphoma, Non-Hodgkin/pathology , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recurrence , Remission Induction , T-Lymphocytes/drug effects , Tissue Donors , Young Adult
5.
Hematol Oncol Stem Cell Ther ; 13(2): 98-105, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32202248

ABSTRACT

Sickle cell disease (SCD) is an inherited disorder; despite significant improvements in supportive care, SCD continues to cause substantial morbidity, mortality, and reduced life expectancy. Allogeneic hematopoietic stem cell transplantation (HSCT) is currently the only widely available curative therapy for SCD, which is offered as a standard of care for patients with a matched sibling donor (MSD). Donor availability is limited to a minority of patients. Thus, αß/CD3-depleted haploidentical HSCT, as an efficient means for depletion of graft-versus-host disease (GvHD)-mediating T cells, can be offered as an alternative curative therapy, particularly for nonmalignant diseases such as SCD. Out of 38 patients with advanced stage SCD, 25 were transplanted with CD3/CD19- or T-cell receptor αß/CD19 T-cell-depleted peripheral stem cell grafts (T-haplo-HSCT group), whereas 13 transplanted from MSD (MSD group); both groups received an almost identical conditioning regimen. Engraftment was achieved in all. However, in the T-haplo-HSCT group, three patients succumbed to an uncontrolled cytomegalovirus pneumonitis, a macrophage activation syndrome, and a major blood group incompatibility with a late graft failure and multiorgan failure. The overall survival was 88% and 100% in T-haplo-HSCT and MSD groups, respectively. None of our patients developed a Glucksberg Grade III-IV acute GvHD. Four patients (16%) in the T-haplo-HSCT group and two patients (15%) in the MSD group developed a steroid-sensitive, mild-to-moderate chronic GvHD that resolved within 18 months posttransplant. These results are encouraging and demonstrate the feasibility, safety, and efficacy of T-haplo-HSCT in advanced stage SCD in children and adults, thus offering a curative alternative to majority of patients.


Subject(s)
Anemia, Sickle Cell/therapy , Antigens, CD19/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , T-Lymphocytes/metabolism , Transplantation Conditioning/methods , Adolescent , Adult , Antigens, CD19/pharmacology , Child , Child, Preschool , Female , Humans , Tissue Donors , Young Adult
6.
J Immunother ; 41(1): 19-31, 2018 01.
Article in English | MEDLINE | ID: mdl-29176334

ABSTRACT

Chimeric antigen receptor (CAR)-based adoptive T-cell therapy is a highly promising treatment for lymphoid malignancies, and CD20 is an ideal target antigen. We previously developed a lentiviral construct encoding a third generation CD20-targeted CAR but identified several features that required additional optimization before clinical translation. We describe here several improvements, including replacement of the immunogenic murine antigen-binding moiety with a fully human domain, streamlining the transgene insert to enhance lentiviral titers, modifications to the extracellular IgG spacer that abrogate nonspecific activation resulting from binding to Fc receptors, and evaluation of CD28, 4-1BB, or CD28 and 4-1BB costimulatory domains. We also found that restimulation of CAR T cells with an irradiated CD20 cell line boosted cell growth, increased the fraction of CAR-expressing cells, and preserved in vivo function despite leading to a reduced capacity for cytokine secretion in vitro. We also found that cryopreservation of CAR T cells did not affect immunophenotype or in vivo antitumor activity compared with fresh cells. These optimization steps resulted in significant improvement in antitumor activity in mouse models, resulting in eradication of established systemic lymphoma tumors in 75% of mice with a single infusion of CAR T cells, and prolonged in vivo persistence of modified cells. These results provide the basis for clinical testing of a lentiviral construct encoding a fully human CD20-targeted CAR with CD28 and 4-1BB costimulatory domains and truncated CD19 (tCD19) transduction marker.


Subject(s)
Immunotherapy, Adoptive/methods , Lymphoma/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/physiology , Animals , Antigens, CD19/pharmacology , Antigens, CD20/immunology , CD28 Antigens/genetics , Cell Culture Techniques , Cells, Cultured , Cytotoxicity, Immunologic , Drug Evaluation, Preclinical , Female , Genetic Engineering , Humans , Lymphocyte Activation , Lymphoma/immunology , Male , Mice , Mice, SCID , Neoplasms, Experimental , Recombinant Fusion Proteins , T-Lymphocytes/transplantation , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Xenograft Model Antitumor Assays
7.
J Immunother ; 28(3): 203-11, 2005.
Article in English | MEDLINE | ID: mdl-15838376

ABSTRACT

Human peripheral blood lymphocytes can be transduced to express antigen-dependent CD3zeta chimeric immune receptors (CIRs), which function independently of the T-cell receptor (TCR). Although the exact function of these domains is unclear, previous studies imply that an extracellular spacer region is required for optimal CIR activity. In this study, four scFvs (in the context of CIRs with or without extracellular spacer regions) were used to target the human tumor-associated antigens carcinoembryonic antigen (CEA), neural cell adhesion molecule (NCAM), the oncofetal antigen 5T4, and the B-cell antigen CD19. In all cases human T-cell populations expressing the CIRs were functionally active against their respective targets, but the anti-5T4 and anti-NCAM CIRs showed enhanced specific cytokine release and cytotoxicity only when possessing an extracellular spacer region. In contrast, the anti-CEA and anti-CD19 CIRs displayed optimal cytokine release activity only in the absence of an extracellular spacer. Interestingly, mapping of the scFv epitopes has revealed that the anti-CEA scFv binds close to the amino-terminal of CEA, which is easily accessible to the CIR. In contrast, CIRs enhanced by a spacer domain appear to bind to epitopes residing closer to the cell membrane, suggesting that a more flexible extracellular domain may be required to permit the efficient binding of such epitopes. These results show that a spacer is not necessary for optimal activity of CIRs but that the optimal design varies.


Subject(s)
Antigens, Neoplasm/immunology , CD3 Complex/genetics , Immunoglobulin Variable Region/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Antigens, CD19/immunology , Antigens, CD19/pharmacology , CD3 Complex/immunology , Carcinoembryonic Antigen/immunology , Carcinoembryonic Antigen/pharmacology , Extracellular Space , Humans , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/pharmacology , Neural Cell Adhesion Molecules/immunology , Neural Cell Adhesion Molecules/pharmacology , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/drug effects , Transfection
8.
Int J Immunopathol Pharmacol ; 18(1): 65-74, 2005.
Article in English | MEDLINE | ID: mdl-15698512

ABSTRACT

With the addition of various cytokines, the CD40-CD40 ligand (CD40L) system can act as a T-helper cell surrogate to permit B lymphocytes to produce large amounts of polyclonal Ig. In the present study, we tested six CD40-CD40L stimulation models: (i, ii) soluble agonistic 89 and G28.5 mAbs ; (iii, iv) 89 and G28.5 bound via their Fc fragments on CDw32-transfected mouse fibroblasts; (v) purified, soluble, trimeric human CD40L molecules (sCD40L); and (vi) human CD40L expressed by a CD40L-transfected mouse fibroblastic cell line (LCD40L). Target B cells consisted of purified blood and tonsillar CD19+ lymphocytes cultured in the presence of CD40 stimuli and IL-2 and IL-10, added at the onset of each B cell culture. A) There was differential expression of CD69, CD80 and CD86 exposure to sCD40L and LCD40L was ensued by the strongest % MFI changes over control. B) In blood B cells, mAbs and sCD40L induced IgA, IgM and IgG production almost equally well; LCD40L proved less efficient. In contrast, in tonsil B cells, LCD40L induced significantly more IgA, IgG1, IgG3 and IgM production than other signals. Using certain CD40/CD40L stimuli to model in vitro Ig production, a system used regularly in many laboratories, may affect the interpretation based on the cell type and on the CD40/CD40L system used.


Subject(s)
Antibodies, Monoclonal/pharmacology , B-Lymphocytes/metabolism , CD40 Antigens/metabolism , Animals , Antigens, CD19/pharmacology , Cell Differentiation/physiology , Cell Membrane/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Flow Cytometry , Humans , Immunoglobulin A/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin M/immunology , Indicators and Reagents , Mice , Palatine Tonsil/cytology , Transfection
10.
J Immunol ; 157(10): 4371-8, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8906812

ABSTRACT

CD19 serves as a cell surface response regulator that establishes signaling thresholds critical for B lymphocyte development and activation. B lymphocytes from CD19-deficient mice are hyporesponsive to transmembrane signals, while B lymphocytes from mice that overexpress CD19 to even a small extent (25% increase) become hyperresponsive. The B-1 subpopulation of B lymphocytes is particularly sensitive to CD19 regulation, since their development is severely decreased in CD19-deficient mice. The effect of altered CD19 expression levels on the development of B cells was therefore examined using transgenic mice that express varying levels of cell surface CD19. In this study, immature B cells within the bone marrow of wild-type mice were found to specifically up-regulate CD19 expression levels by twofold as they mature, while CD5+ B cells within the spleen and peritoneum expressed even higher levels of CD19. The development of CD5+ B cells was severely decreased in CD19-deficient mice, while there was a linear correlation between increased CD19 expression levels and the increased frequency of CD5+ B cells within the peritoneum and spleen. In fact, CD5+ B cells became a major B lymphocyte population in mice that overexpressed CD19. Increased expression of CD19 also correlated with increased levels of endogenous anti-DNA Abs and rheumatoid factor. These results indicate that up-regulated expression of CD19 is functionally important for B cell development and that CD19 establishes signaling thresholds that regulate the generation of B-1 lymphocytes as well as the development of autoantibodies.


Subject(s)
Antigens, CD19/pharmacology , Autoimmunity/drug effects , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Signal Transduction/immunology , Animals , B-Lymphocyte Subsets/drug effects , Cell Differentiation/drug effects , Cell Differentiation/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred NZB , Mice, Mutant Strains , Mice, Transgenic , Signal Transduction/drug effects
11.
Leuk Lymphoma ; 18(1-2): 93-102, 1995 Jun.
Article in English | MEDLINE | ID: mdl-8580835

ABSTRACT

Standard immunotoxin production procedures using whole IgG as the MoAb moiety yield a heterogeneous mixture of 180 kDa, 210 kDa, and 240 kDa immunotoxin species with 1 to 1, 1 to 2, and 1 to 3 MoAb to toxin ratios. This heterogeneity makes it impossible to precisely deliver a predetermined immunotoxin dose to target cells and impairs the accuracy of pharmacologic studies. In this report, we describe the preparation and characterization of B43(anti-CD19)-pokeweed antiviral protein (PAP) immunotoxins containing either one or two 30 kDa PAP toxin molecules covalently linked to each 150 kDa B43 monoclonal antibody molecule. Compared to the 180 kDa immunotoxin, the 210 kDa immunotoxin displayed greater in vitro chemical stability, resulted in higher systemic exposure levels in vivo, and was a more effective anti-leukemic agent in a SCID mouse model of human B-lineage acute lymphoblastic leukemia. Taken together, the results of this study recommend the clinical evaluation of 210 kDa B43-PAP as a potentially more effective immunotoxin against relapsed B-lineage ALL.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antiviral Agents/pharmacology , Burkitt Lymphoma/drug therapy , Immunotoxins/pharmacology , N-Glycosyl Hydrolases , Plant Proteins/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antigens, CD19/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antiviral Agents/pharmacokinetics , Burkitt Lymphoma/metabolism , Drug Stability , Female , Immunotoxins/chemistry , Immunotoxins/pharmacokinetics , Mice , Mice, Inbred BALB C , Mice, SCID , Plant Proteins/chemistry , Plant Proteins/pharmacokinetics , Rabbits , Ribosome Inactivating Proteins, Type 1
SELECTION OF CITATIONS
SEARCH DETAIL
...