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










Publication year range
1.
Mol Ther Methods Clin Dev ; 22: 377-387, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34514029

ABSTRACT

With the US Food and Drug Administration (FDA) approval of four CD19- and one BCMA-targeted chimeric antigen receptor (CAR) therapy for B cell malignancies, CAR T cell therapy has finally reached the status of a medicinal product. The successful manufacturing of autologous CAR T cell products is a key requirement for this promising treatment modality. By analyzing the composition of 214 apheresis products from 210 subjects across eight disease indications, we found that high CD14+ cell content poses a challenge for manufacturing CAR T cells, especially in patients with non-Hodgkin's lymphoma and multiple myeloma caused by the non-specific phagocytosis of the magnetic beads used to activate CD3+ T cells. We demonstrated that monocyte depletion via rapid plastic surface adhesion significantly reduces the CD14+ monocyte content in the apheresis products and simultaneously boosts the CD3+ content. We established a 40% CD14+ threshold for the stratification of apheresis products across nine clinical trials and demonstrated the effectiveness of this procedure by comparing manufacturing runs in two phase 1 clinical trials. Our study suggests that CD14+ content should be monitored in apheresis products, and that the manufacturing of CAR T cells should incorporate a step that lessens the CD14+ cell content in apheresis products containing more than 40% to maximize the production success.

2.
Sci Transl Med ; 6(224): 224ra25, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-24553386

ABSTRACT

We report on 16 patients with relapsed or refractory B cell acute lymphoblastic leukemia (B-ALL) that we treated with autologous T cells expressing the 19-28z chimeric antigen receptor (CAR) specific to the CD19 antigen. The overall complete response rate was 88%, which allowed us to transition most of these patients to a standard-of-care allogeneic hematopoietic stem cell transplant (allo-SCT). This therapy was as effective in high-risk patients with Philadelphia chromosome-positive (Ph(+)) disease as in those with relapsed disease after previous allo-SCT. Through systematic analysis of clinical data and serum cytokine levels over the first 21 days after T cell infusion, we have defined diagnostic criteria for a severe cytokine release syndrome (sCRS), with the goal of better identifying the subset of patients who will likely require therapeutic intervention with corticosteroids or interleukin-6 receptor blockade to curb the sCRS. Additionally, we found that serum C-reactive protein, a readily available laboratory study, can serve as a reliable indicator for the severity of the CRS. Together, our data provide strong support for conducting a multicenter phase 2 study to further evaluate 19-28z CAR T cells in B-ALL and a road map for patient management at centers now contemplating the use of CAR T cell therapy.


Subject(s)
Cell Transplantation , Immunotherapy , Leukemia, B-Cell/therapy , T-Lymphocytes/immunology , Adolescent , Adult , Female , Humans , Male , Middle Aged , Young Adult
3.
Sci Transl Med ; 5(177): 177ra38, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23515080

ABSTRACT

Adults with relapsed B cell acute lymphoblastic leukemia (B-ALL) have a dismal prognosis. Only those patients able to achieve a second remission with no minimal residual disease (MRD) have a hope for long-term survival in the context of a subsequent allogeneic hematopoietic stem cell transplantation (allo-HSCT). We have treated five relapsed B-ALL subjects with autologous T cells expressing a CD19-specific CD28/CD3ζ second-generation dual-signaling chimeric antigen receptor (CAR) termed 19-28z. All patients with persistent morphological disease or MRD(+) disease upon T cell infusion demonstrated rapid tumor eradication and achieved MRD(-) complete remissions as assessed by deep sequencing polymerase chain reaction. Therapy was well tolerated, although significant cytokine elevations, specifically observed in those patients with morphologic evidence of disease at the time of treatment, required lymphotoxic steroid therapy to ameliorate cytokine-mediated toxicities. Indeed, cytokine elevations directly correlated to tumor burden at the time of CAR-modified T cell infusions. Tumor cells from one patient with relapsed disease after CAR-modified T cell therapy, who was ineligible for additional allo-HSCT or T cell therapy, exhibited persistent expression of CD19 and sensitivity to autologous 19-28z T cell-mediated cytotoxicity, which suggests potential clinical benefit of additional CAR-modified T cell infusions. These results demonstrate the marked antitumor efficacy of 19-28z CAR-modified T cells in patients with relapsed/refractory B-ALL and the reliability of this therapy to induce profound molecular remissions, forming a highly effective bridge to potentially curative therapy with subsequent allo-HSCT.


Subject(s)
Antigens, CD19/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Adult , Aged , Cells, Cultured , Female , Humans , Immunotherapy/methods , Male , Middle Aged , Treatment Outcome , Young Adult
4.
Blood ; 118(18): 4817-28, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-21849486

ABSTRACT

We report the findings from the first 10 patients with chemotherapy-refractory chronic lymphocytic leukemia (CLL) or relapsed B-cell acute lymphoblastic leukemia (ALL) we have enrolled for treatment with autologous T cells modified to express 19-28z, a second-generation chimeric antigen (Ag) receptor specific to the B-cell lineage Ag CD19. Eight of the 9 treated patients tolerated 19-28z(+) T-cell infusions well. Three of 4 evaluable patients with bulky CLL who received prior conditioning with cyclophosphamide exhibited either a significant reduction or a mixed response in lymphadenopathy without concomitant development of B-cell aplasia. In contrast, one patient with relapsed ALL who was treated in remission with a similar T-cell dose developed a predicted B-cell aplasia. The short-term persistence of infused T cells was enhanced by prior cyclophosphamide administration and inversely proportional to the peripheral blood tumor burden. Further analyses showed rapid trafficking of modified T cells to tumor and retained ex vivo cytotoxic potential of CD19-targeted T cells retrieved 8 days after infusion. We conclude that this adoptive T-cell approach is promising and more likely to show clinical benefit in the setting of prior conditioning chemotherapy and low tumor burden or minimal residual disease. These studies are registered at www.clinicaltrials.org as #NCT00466531 (CLL protocol) and #NCT01044069 (B-ALL protocol).


Subject(s)
Antigens, CD19/immunology , Graft Survival , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Leukemia, B-Cell/therapy , T-Lymphocytes/transplantation , Adult , Aged , Antigens, CD19/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/physiology , Female , Graft Survival/physiology , Humans , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/immunology , Male , Middle Aged , Recurrence , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Transplantation, Autologous , Treatment Failure
5.
J Immunother ; 32(2): 169-80, 2009.
Article in English | MEDLINE | ID: mdl-19238016

ABSTRACT

On the basis of promising preclinical data demonstrating the eradication of systemic B-cell malignancies by CD19-targeted T lymphocytes in vivo in severe combined immunodeficient-beige mouse models, we are launching phase I clinical trials in patients with chronic lymphocytic leukemia (CLL) and acute lymphoblastic leukemia. We present here the validation of the bioprocess which we developed for the production and expansion of clinical grade autologous T cells derived from patients with CLL. We demonstrate that T cells genetically modified with a replication-defective gammaretroviral vector derived from the Moloney murine leukemia virus encoding a chimeric antigen receptor (CAR) targeted to CD19 (1928z) can be expanded with Dynabeads CD3/CD28. This bioprocess allows us to generate clinical doses of 1928z+ T cells in approximately 2 to 3 weeks in a large-scale semiclosed culture system using the Wave Bioreactor. These 1928z+ T cells remain biologically functional not only in vitro but also in severe combined immunodeficient-beige mice bearing disseminated tumors. The validation requirements in terms of T-cell expansion, T-cell transduction with the 1928z CAR, biologic activity, quality control testing, and release criteria were met for all 4 validation runs using apheresis products from patients with CLL. Additionally, after expansion of the T cells, the diversity of the skewed Vbeta T-cell receptor repertoire was significantly restored. This validated process will be used in phase I clinical trials in patients with chemorefractory CLL and in patients with relapsed acute lymphoblastic leukemia. It can also be adapted for other clinical trials involving the expansion and transduction of patient or donor T cells using any CAR or T-cell receptor.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Receptors, Antigen/genetics , T-Lymphocytes, Cytotoxic/immunology , Animals , Bioreactors , Cell Culture Techniques , Clinical Trials as Topic , Genetic Engineering , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Mice , Receptors, Antigen/immunology , T-Lymphocytes, Cytotoxic/transplantation , Transduction, Genetic
6.
Vaccine ; 25(27): 5013-24, 2007 Jun 28.
Article in English | MEDLINE | ID: mdl-17537555

ABSTRACT

The use of plasmid DNA as vaccines for the treatment of cancer and infectious diseases is on the rise. In order to facilitate the manufacture of clinical-grade plasmid DNA for Phase I clinical trials, we developed a process whereby >200 mg plasmid could be produced in a single production run under Good Manufacturing Practices. A dedicated cleanroom (Class 10,000 with Class 100 biosafety cabinet) is utilized for production of the bacterial cell bank, fermentation, harvest/lysis of the biomass, and downstream purification. Fermentation requires three 16-18 h runs (approximately 12 L each) in shaker-flasks, yielding approximately 60 g bacterial paste following batch centrifugation. The biomass is alkaline-lysed, pooled, and the resulting flocculent precipitate is separated by a novel vacuum step, followed by depth-filtration. Downstream processing includes anion-exchange chromatography, utilizing Qiagen silica-based resin, and precipitation with isopropanol. Following precipitation, the DNA is harvested by centrifugation, dried, formulated, and sterile-filtered using a Sartorius Sartobran 150 filter prior to Final-Filling. All processing steps utilize sterilized, single-use components. This process results in a product manufactured according to regulatory guidelines. The plasmid DNA is sterile with >or=95% supercoiled DNA, an A260/A280 ratio>or=1.9, undetectable or extremely low residual endotoxin, RNA, genomic DNA, protein, and antibiotic. Residual solvent levels are negligible. The product yields the predicted profile upon restriction-enzyme digestion, is biologically active upon transfection and remains stable for several years at -20 degrees C. We have therefore developed a reproducible and cost effective process to manufacture clinical-grade plasmid DNA. This process can be adapted by other academic centers for human or large animal clinical trials.


Subject(s)
Clinical Trials, Phase I as Topic/standards , DNA/biosynthesis , Drug Industry , Plasmids/genetics , Vaccines, DNA/biosynthesis , Animals , Cellulose/analogs & derivatives , Chromatography, Ion Exchange , Chromosome Mapping , Culture Media , DNA/analysis , Electrophoresis, Polyacrylamide Gel , Fermentation , Humans , Indicators and Reagents , Vaccines, DNA/analysis
7.
Proc Natl Acad Sci U S A ; 102(14): 5044-9, 2005 Apr 05.
Article in English | MEDLINE | ID: mdl-15795385

ABSTRACT

Reactive oxygen species (ROS) are key components of postreceptor intracellular signaling pathways; however, the role of ROS in signal initiation is uncertain. We discovered that receptor-ligand interaction caused the generation of hydrogen peroxide (H2O2). Using members of the hematopoietin receptor superfamily, as well as EGF receptor, we show that H2O2 is generated by specific receptor-ligand interaction in cells and in cell-free systems. With cognate ligand, the extracellular domain of the receptor was sufficient for H2O2 generation. We also found that production of H2O2 was diminished in a granulocyte-macrophage colony-stimulating factor receptor mutant unable to bind ligand. Exogenously added H2O2 induced signaling in the absence of ligand, whereas catalase and a membrane-bound peroxiredoxin inhibited ligand-dependent signaling. Our results suggest that H2O2 produced by receptor-ligand interaction is involved as a chemical mediator that facilitates cell signaling.


Subject(s)
Hydrogen Peroxide/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Base Sequence , Catalase/pharmacology , Cell Line , DNA, Complementary/genetics , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Extracellular Fluid/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , In Vitro Techniques , Ligands , Models, Biological , Mutation , Peroxidases/genetics , Peroxidases/metabolism , Peroxiredoxins , Protein Subunits , Reactive Oxygen Species/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
8.
Mol Cell Biol ; 24(15): 6645-52, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15254232

ABSTRACT

Reactive oxygen species (ROS) are key intermediates in cellular signal transduction pathways whose function may be counterbalanced by antioxidants. Acting as an antioxidant, ascorbic acid (AA) donates two electrons and becomes oxidized to dehydroascorbic acid (DHA). We discovered that DHA directly inhibits IkappaBalpha kinase beta (IKKbeta) and IKKalpha enzymatic activity in vitro, whereas AA did not have this effect. When cells were loaded with AA and induced to generate DHA by oxidative stress in cells expressing a constitutive active IKKbeta, NF-kappaB activation was inhibited. Our results identify a dual molecular action of vitamin C in signal transduction and provide a direct linkage between the redox state of vitamin C and NF-kappaB signaling events. AA quenches ROS intermediates involved in the activation of NF-kappaB and is oxidized to DHA, which directly inhibits IKKbeta and IKKalpha enzymatic activity. These findings define a function for vitamin C in signal transduction other than as an antioxidant and mechanistically illuminate how vitamin C down-modulates NF-kappaB signaling.


Subject(s)
Ascorbic Acid/pharmacology , Dehydroascorbic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Antioxidants/metabolism , Ascorbic Acid/metabolism , DNA/metabolism , Dose-Response Relationship, Drug , Down-Regulation , HeLa Cells , Humans , I-kappa B Kinase , Immunoblotting , Luciferases/metabolism , Models, Biological , Oxidative Stress , Oxygen/metabolism , Protein Binding , Reactive Oxygen Species , Signal Transduction , Transfection
9.
Proc Natl Acad Sci U S A ; 100(24): 14000-5, 2003 Nov 25.
Article in English | MEDLINE | ID: mdl-14614142

ABSTRACT

Basement membrane matrix proteins are known to up-regulate granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling in neutrophils and mononuclear phagocytes, but the mechanisms involved are poorly understood. We used the intracellular portion of the alpha subunit of the GM-CSF receptor (alphaGMR) to search for interacting proteins and identified the 67-kDa laminin receptor (LR), a nonintegrin matrix protein receptor expressed in several types of host defense cells and certain tumors, as a binding partner. LR was found to interact with the beta subunit of the GMR (betaGMR) as well. Whereas GM-CSF functions by engaging the alphaGMR and betaGMR into receptor complexes, LR inhibited GM-CSF-induced receptor complex formation. Laminin and fibronectin binding to LR was found to prevent the binding of betaGMR to LR and relieved the LR inhibition of GMR. These findings provide a mechanistic basis for enhancing host defense cell responsiveness to GM-CSF at transendothelial migration sites while suppressing it in circulation.


Subject(s)
Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Laminin/metabolism , Base Sequence , Binding Sites , Cell Line , DNA, Complementary/genetics , Fibronectins/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , In Vitro Techniques , Kinetics , Laminin/metabolism , Macromolecular Substances , Models, Molecular , Molecular Weight , Neutrophils/metabolism , Protein Structure, Tertiary , Protein Subunits , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Laminin/chemistry , Signal Transduction
10.
Biochemistry ; 41(43): 12995-3002, 2002 Oct 29.
Article in English | MEDLINE | ID: mdl-12390026

ABSTRACT

Extracellular stimuli signal for activation of the transcription factor NFkappaB, leading to gene expression regulating processes involved in immune responses, inflammation, and cell survival. Tumor necrosis factor-alpha (TNFalpha) activates NFkappaB via a well-defined kinase pathways involving NFkappaB-inducing kinase (NIK), which activates downstream multisubunit IkappaB kinases (IKK). IKK in turn phosphorylates IkappaB, the central regulator of NFkappaB function. We found that intracellular vitamin C inhibits TNFalpha-induced activation of NFkappaB in human cell lines (HeLa, monocytic U937, myeloid leukemia HL-60, and breast MCF7) and primary endothelial cells (HUVEC) in a dose-dependent manner. Vitamin C is an important antioxidant, and most cells accumulate ascorbic acid (AA) intracellularly by transporting the oxidized form of the vitamin, dehydroascorbic acid (DHA). Because ascorbic acid is a strong pro-oxidant in the presence of transition metals in vitro, we loaded cells with vitamin C by incubating them with DHA. Vitamin C-loaded cells showed significantly decreased TNFalpha-induced nuclear translocation of NFkappaB, NFkappaB-dependent reporter transcription, and IkappaBalpha phosphorylation. Our data point to a mechanism of vitamin C suppression of NFkappaB activation by inhibiting TNFalpha-induced activation of NIK and IKKbeta kinases independent of p38 MAP kinase. These results suggest that intracellular vitamin C can influence inflammatory, neoplastic, and apoptotic processes via inhibition of NFkappaB activation.


Subject(s)
Ascorbic Acid/pharmacology , I-kappa B Proteins/antagonists & inhibitors , I-kappa B Proteins/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , HL-60 Cells , HeLa Cells , Humans , I-kappa B Kinase , Mitogen-Activated Protein Kinases/metabolism , NF-KappaB Inhibitor alpha , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Transcriptional Activation/drug effects , U937 Cells , p38 Mitogen-Activated Protein Kinases , NF-kappaB-Inducing Kinase
11.
Blood ; 99(9): 3205-12, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11964284

ABSTRACT

Vitamin C is present in the cytosol as ascorbic acid, functioning primarily as a cofactor for enzymatic reactions and as an antioxidant to scavenge free radicals. Human granulocyte macrophage-colony-stimulating factor (GM-CSF) induces an increase in reactive oxygen species (ROS) and uses ROS for some signaling functions. We therefore investigated the effect of vitamin C on GM-CSF-mediated responses. Loading U937 cells with vitamin C decreased intracellular levels of ROS and inhibited the production of ROS induced by GM-CSF. Vitamin C suppressed GM-CSF-dependent phosphorylation of the signal transducer and activator of transcription 5 (Stat-5) and mitogen-activated protein (MAP) kinase (Erk1 and Erk2) in a dose-dependent manner as was phosphorylation of MAP kinase induced by both interleukin 3 (IL-3) and GM-CSF in HL-60 cells. In 293T cells transfected with alpha and beta GM-CSF receptor subunits (alphaGMR and betaGMR), GM-CSF-induced phosphorylation of betaGMR and Jak-2 activation was suppressed by vitamin C loading. GM-CSF-mediated transcriptional activation of a luciferase reporter construct containing STAT-binding sites was also inhibited by vitamin C. These results substantiate the importance of ROS in GM-CSF signaling and indicate a role for vitamin C in downmodulating GM-CSF signaling responses. Our findings point to vitamin C as a regulator of cytokine redox-signal transduction in host defense cells and a possible role in controlling inflammatory responses.


Subject(s)
Ascorbic Acid/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Milk Proteins , Signal Transduction/drug effects , Adjuvants, Immunologic/pharmacology , Cell Division/drug effects , DNA-Binding Proteins/metabolism , Drug Antagonism , Homeostasis , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , STAT5 Transcription Factor , Trans-Activators/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...