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










Publication year range
1.
Cancer Immunol Immunother ; 73(3): 60, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38400933

ABSTRACT

Over the past decade, US Food and Drug Administration (FDA)-approved immune checkpoint inhibitors that target programmed death-1 (PD-1) have demonstrated significant clinical benefit particularly in patients with PD-L1 expressing tumors. Toripalimab is a humanized anti-PD-1 antibody, approved by FDA for first-line treatment of nasopharyngeal carcinoma in combination with chemotherapy. In a post hoc analysis of phase 3 studies, toripalimab in combination with chemotherapy improved overall survival irrespective of PD-L1 status in nasopharyngeal carcinoma (JUPITER-02), advanced non-small cell lung cancer (CHOICE-01) and advanced esophageal squamous cell carcinoma (JUPITER-06). On further characterization, we determined that toripalimab is molecularly and functionally differentiated from pembrolizumab, an anti-PD-1 mAb approved previously for treating a wide spectrum of tumors. Toripalimab, which binds the FG loop of PD-1, has 12-fold higher binding affinity to PD-1 than pembrolizumab and promotes significantly more Th1- and myeloid-derived inflammatory cytokine responses in healthy human PBMCs in vitro. In an ex vivo system employing dissociated tumor cells from treatment naïve non-small cell lung cancer patients, toripalimab induced several unique genes in IFN-γ and immune cell pathways, showed different kinetics of activation and significantly enhanced IFN-γ signature. Additionally, binding of toripalimab to PD-1 induced lower levels of SHP1 and SHP2 recruitment, the negative regulators of T cell activation, in Jurkat T cells ectopically expressing PD-1. Taken together, these data demonstrate that toripalimab is a potent anti-PD-1 antibody with high affinity PD-1 binding, strong functional attributes and demonstrated clinical activity that encourage its continued clinical investigation in several types of cancer.


Subject(s)
Antibodies, Monoclonal, Humanized , Carcinoma, Non-Small-Cell Lung , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Lung Neoplasms , Nasopharyngeal Neoplasms , Humans , Antibodies, Monoclonal/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , B7-H1 Antigen , Programmed Cell Death 1 Receptor , Lung Neoplasms/drug therapy , Nasopharyngeal Carcinoma , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , T-Lymphocytes/pathology
2.
Adv Ther ; 39(3): 1230-1246, 2022 03.
Article in English | MEDLINE | ID: mdl-35034311

ABSTRACT

INTRODUCTION: Biologic therapeutics can trigger immune responses in patients. As part of the totality of evidence that is required for regulatory approval of biosimilars, immunogenicity similarity must be assessed in the clinical programs. Pegfilgrastim-cbqv (UDENYCA®) is a pegfilgrastim biosimilar approved in the USA and European Union. This article demonstrates the similar immunogenicity of pegfilgrastim-cbqv compared with its reference product, pegfilgrastim (Neulasta®). METHODS: The immunogenicity of pegfilgrastim-cbqv was assessed in three clinical studies in healthy subjects (one specifically designed to evaluate immunogenicity similarity and two studies to assess pharmacokinetics and pharmacodynamics bioequivalence) using a tiered approach, in which plasma samples were tested for the presence of antidrug antibodies (ADAs) as well as ADA binding-specificity, titer and neutralizing activity. To assess the clinical impact of ADAs, pharmacokinetics, pharmacodynamics and safety profiles were compared between ADA-positive and -negative subjects. RESULTS: These studies demonstrated similar immunogenicity of pegfilgrastim-cbqv and pegfilgrastim. The small differences in ADA incidence between treatment groups observed in the immunogenicity study were driven by non-neutralizing, low-titer, polyethylene glycol (PEG)-reactive ADAs, which are commonly present in healthy subjects. No treatment-emergent neutralizing antibodies (NAbs) were detected in either treatment group, and there was no apparent impact of ADAs on pharmacokinetics, pharmacodynamics or safety. CONCLUSION: Pegfilgrastim-cbqv has similar immunogenicity to pegfilgrastim. The presented immunogenicity, pharmacokinetics, pharmacodynamics and safety data support the overall demonstration of no clinically meaningful differences between pegfilgrastim-cbqv and pegfilgrastim. CLINICAL TRIAL REGISTRATION: NCT02418104 (CHS-1701-04, April 2015), NCT02650973 (CHS-1701-05, February 2016) and NCT02385851 (CHS-1701-03, March 2015).


Subject(s)
Biosimilar Pharmaceuticals , Biosimilar Pharmaceuticals/pharmacology , Biosimilar Pharmaceuticals/therapeutic use , Case-Control Studies , Filgrastim/therapeutic use , Healthy Volunteers , Humans , Polyethylene Glycols/pharmacology
3.
Adv Ther ; 37(10): 4291-4307, 2020 10.
Article in English | MEDLINE | ID: mdl-32789809

ABSTRACT

INTRODUCTION: Pegfilgrastim-cbqv was developed as a biosimilar of pegfilgrastim, a pegylated form of recombinant human granulocyte colony-stimulating factor approved for decreasing febrile neutropenia-associated infection in patients receiving myelosuppressive drugs. This multicenter, randomized, single-blind, partial-reference-replicated, three-sequence crossover study assessed pharmacokinetic and pharmacodynamic bioequivalence of pegfilgrastim-cbqv and pegfilgrastim in healthy subjects. METHODS: One hundred twenty-two subjects were randomized to one of three treatment sequences; each included one dose of pegfilgrastim-cbqv and two doses of pegfilgrastim separated by ≥ 28 days. The primary pharmacokinetic end points were area under the curve (AUC) from 0 to infinity (AUC0-∞) and maximum concentration (Cmax). The primary pharmacodynamic end points were maximum absolute neutrophil count (ANCmax) and ANC AUC from time 0 to the last measurable observation (ANC AUC0-last). Pharmacokinetic and pharmacodynamic bioequivalences were demonstrated if the 90% CI for the geometric mean ratio (GMR) of pegfilgrastim-cbqv to pegfilgrastim was within 80-125% for the primary end points. RESULTS: Pharmacokinetic bioequivalence criteria were met for Cmax (GMR 105.0; 90% CI 95.5-115.4) and AUC0-∞ (GMR 97.5; 90% CI 88.6-107.2). Pharmacodynamic bioequivalence criteria were met for ANCmax (GMR 99.6; 90% CI 96.2-103.2) and ANC AUC0-last (GMR 96.7; 90% CI 92.2-101.4). Adverse events occurred in 76.0%, 76.6%, and 73.1% of subjects for pegfilgrastim-cbqv, first pegfilgrastim, and second pegfilgrastim dosing periods across treatment sequences, respectively. Investigators found no drug-related serious adverse events. CONCLUSION: This study established pharmacokinetic and pharmacodynamic bioequivalence of pegfilgrastim-cbqv to pegfilgrastim. The treatments displayed similar safety profiles, including immunogenicity, with no unexpected safety findings. CLINICAL TRIALS REGISTRATION: ClinicalTrials.gov, NCT02650973, February 2016.


Subject(s)
Granulocyte Colony-Stimulating Factor , Polyethylene Glycols , Cross-Over Studies , Filgrastim , Healthy Volunteers , Humans , Recombinant Proteins , Single-Blind Method
4.
MAbs ; 8(7): 1417-1424, 2016 10.
Article in English | MEDLINE | ID: mdl-27367933

ABSTRACT

The CD25-binding antibody daclizumab high-yield process (DAC HYP) is an interleukin (IL)-2 signal modulating antibody that shares primary amino acid sequence and CD25 binding affinity with Zenapax®, a distinct form of daclizumab, which was approved for the prevention of acute organ rejection in patients receiving renal transplants as part of an immunosuppressive regimen that includes cyclosporine and corticosteroids. Comparison of the physicochemical properties of the two antibody forms revealed the glycosylation profile of DAC HYP differs from Zenapax in both glycan distribution and the types of oligosaccharides, most notably high-mannose, galactosylated and galactose-α-1,3-galactose (α-Gal) oligosaccharides, resulting in a DAC HYP antibody material that is structurally distinct from Zenapax. Although neither antibody elicited complement-dependent cytotoxicity in vitro, DAC HYP antibody had significantly reduced levels of antibody-dependent cell-mediated cytotoxicity (ADCC). The ADCC activity required natural killer (NK) cells, but not monocytes, suggesting the effects were mediated through binding to Fc-gamma RIII (CD16). Incubation of each antibody with peripheral blood mononuclear cells also caused the down-modulation of CD16 expression on NK cells and the CD16 down-modulation was greater for Zenapax in comparison to that observed for DAC HYP. The substantive glycosylation differences between the two antibody forms and corresponding greater Fc-mediated effector activities by Zenapax, including cell killing activity, manifest as a difference in the biological function and pharmacology between DAC HYP and Zenapax.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Immunoglobulin G/pharmacology , Leukocytes, Mononuclear/drug effects , Daclizumab , Glycosylation , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Leukocytes, Mononuclear/immunology
5.
Arthritis Res Ther ; 15(6): R207, 2013.
Article in English | MEDLINE | ID: mdl-24299175

ABSTRACT

INTRODUCTION: Targeting the CD20 antigen has been a successful therapeutic intervention in the treatment of rheumatoid arthritis (RA). However, in some patients with an inadequate response to anti-CD20 therapy, a persistence of CD20- plasmablasts is noted. The strong expression of CD319 on CD20- plasmablast and plasma cell populations in RA synovium led to the investigation of the potential of CD319 as a therapeutic target. METHODS: PDL241, a novel humanized IgG1 monoclonal antibody (mAb) to CD319, was generated and examined for its ability to inhibit immunoglobulin production from plasmablasts and plasma cells generated from peripheral blood mononuclear cells (PBMC) in the presence and absence of RA synovial fibroblasts (RA-SF). The in vivo activity of PDL241 was determined in a human PBMC transfer into NOD scid IL-2 gamma chain knockout (NSG) mouse model. Finally, the ability of PDL241 to ameliorate experimental arthritis was evaluated in a collagen-induced arthritis (CIA) model in rhesus monkeys. RESULTS: PDL241 bound to plasmablasts and plasma cells but not naïve B cells. Consistent with the binding profile, PDL241 inhibited the production of IgM from in vitro PBMC cultures by the depletion of CD319+ plasmablasts and plasma cells but not B cells. The activity of PDL241 was dependent on an intact Fc portion of the IgG1 and mediated predominantly by natural killer cells. Inhibition of IgM production was also observed in the human PBMC transfer to NSG mouse model. Treatment of rhesus monkeys in a CIA model with PDL241 led to a significant inhibition of anti-collagen IgG and IgM antibodies. A beneficial effect on joint related parameters, including bone remodeling, histopathology, and joint swelling was also observed. CONCLUSIONS: The activity of PDL241 in both in vitro and in vivo models highlights the potential of CD319 as a therapeutic target in RA.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibody Formation/drug effects , Arthritis, Rheumatoid/immunology , Plasma Cells/immunology , Receptors, Immunologic/immunology , Animals , Flow Cytometry , Heterografts , Humans , Immunoglobulins/biosynthesis , Immunohistochemistry , Macaca mulatta , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Signaling Lymphocytic Activation Molecule Family , Synovial Membrane/immunology , Synovial Membrane/metabolism
6.
MAbs ; 5(5): 776-86, 2013.
Article in English | MEDLINE | ID: mdl-23924796

ABSTRACT

CD81 is an essential receptor for hepatitis C virus (HCV). K21 is a novel high affinity anti-CD81 antibody with potent broad spectrum anti-HCV activity in vitro. The pharmacokinetics (PK), pharmacodynamics and liver distribution of K21 were characterized in cynomolgus monkeys after intravenous (i.v.) administration of K21. Characteristic target-mediated drug disposition (TMDD) was shown based on the PK profile of K21 and a semi-mechanistic TMDD model was used to analyze the data. From the TMDD model, the estimated size of the total target pool at baseline (V(c) • R(base)) is 16 nmol/kg and the estimated apparent Michaelis-Menten constant (KM) is 4.01 nM. A simulation using estimated TMDD parameters indicated that the number of free receptors remains below 1% for at least 3 h after an i.v. bolus of 7 mg/kg. Experimentally, the availability of free CD81 on peripheral lymphocytes was measured by immunostaining with anti-CD81 antibody JS81. After K21 administration, a dose- and time-dependent reduction in free CD81 on peripheral lymphocytes was observed. Fewer than 3% of B cells could bind JS81 3 h after a 7 mg/kg dose. High concentrations of K21 were found in liver homogenates, and the liver/serum ratio of K21 increased time-dependently and reached ~160 at 168 h post-administration. The presence of K21 bound to hepatocytes was confirmed by immunohistochemistry. The fast serum clearance of K21 and accumulation in the liver are consistent with TMDD. The TMDD-driven liver accumulation of the anti-CD81 antibody K21 supports the further investigation of K21 as a therapeutic inhibitor of HCV entry.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Liver/metabolism , Models, Biological , Tetraspanin 28/antagonists & inhibitors , Administration, Intravenous , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/immunology , Antibody Affinity/immunology , Area Under Curve , CHO Cells , Cells, Cultured , Computer Simulation , Cricetinae , Cricetulus , Drug Delivery Systems/methods , Hepatocytes/immunology , Hepatocytes/metabolism , Humans , Kinetics , Liver/cytology , Liver/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Macaca fascicularis , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Microscopy, Confocal , Tetraspanin 28/immunology , Tetraspanin 28/metabolism , Time Factors
7.
Mult Scler ; 17(12): 1441-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21807759

ABSTRACT

OBJECTIVE: The objective of this study was to evaluate whether interleukin-2 (IL-2) receptor expression on CD56(bright) natural killer (NK) cells predicted CD56(bright) NK cell expansion and therapeutic response to daclizumab (DAC) in multiple sclerosis (MS). METHODS: DAC exposure, CD56(bright) NK cell counts, IL-2 receptor alpha (CD25) and beta (CD122) subunits, and new or enlarged lesions on brain MRI were measured in 64 subjects in a pharmacokinetic/pharmacodynamic substudy of the phase 2 CHOICE trial at multiple time points. Peripheral blood mononuclear cell (PBMC) samples were obtained from healthy subjects to assess the relationship among DAC treatment, intermediate affinity IL-2 signaling, and CD56(bright) NK cell expansion. RESULTS: Increased CD56(bright) NK cell counts in DAC/interferon beta (IFNß)-treated subjects were observed by day 14, the first post-dosing time point (mean [SD] ln{CD56(bright) NK cell count}: DAC high/IFNß, 2.01 [1.25]; DAC low/IFNß, 2.29 [1.06]; placebo/IFNß, 1.01 [1.03]; adjusted p = 0.003), and persisted throughout the treatment period. Higher DAC dose predicted a faster rate of CD56(bright) NK cell expansion (p < 0.001), but individual subjects' increases in CD56(bright) NK cells from baseline levels were only weakly correlated with DAC exposure (r(2) = 0.167). Higher expression of the intermediate-affinity IL-2 receptor subunit (CD122) on CD56(bright) NK cells at baseline predicted fewer new gadolinium-enhanced (Gd+) lesions during the treatment period (1.77 vs. 0.62 adjusted mean new Gd+ lesions during weeks 8-24, lowest vs. highest quartile of percentage CD122(+) CD56(bright) NK cells; p = 0.033) and a greater increase in CD56(bright) NK cell counts at the end of DAC dosing (p = 0.029). CONCLUSION: CD56(bright) NK cell expansion after DAC treatment appears to reflect individual differences in the capacity for intermediate-affinity IL-2 signaling and could provide a basis for predicting clinical response to DAC in MS.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , CD56 Antigen/analysis , Immunoglobulin G/therapeutic use , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Killer Cells, Natural/immunology , Multiple Sclerosis/immunology , Cell Proliferation/drug effects , Daclizumab , Humans , Killer Cells, Natural/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism
8.
J Nucl Med ; 50(10): 1683-91, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19759100

ABSTRACT

UNLABELLED: Visilizumab is an IgG(2) humanized monoclonal antibody (mAb) characterized by non-FcgammaR binding and specific to the CD3 antigen, expressed on more than 95% of circulating resting T-lymphocytes and on activated T-lymphocytes homing in inflamed tissues. We hypothesized that the use of a radiolabeled anti-CD3 antibody might serve as a diagnostic tool for imaging T-cell traffic and lymphocytic infiltration of tissues and organs affected by autoimmune diseases. Here we describe the results of in vitro and animal experiments with (99m)Tc-succinimidyl-6-hydrazinonicotinate hydrochloride (SHNH)-visilizumab. METHODS: For mAb labeling, we used a 2-step method with a heterobifunctional linker SHNH. Several titrations were performed to obtain the best labeling efficiency. In vitro quality controls included stability assay, cysteine challenge, sodium dodecyl sulfate polyacrylamide gel electrophoresis, binding assay, and immunoreactivity assay. In vivo studies by high-resolution images were performed at 6 and 24 h after the injection of (99m)Tc-SHNH-visilizumab. These included cell-targeting experiments in BALB/c mice xenografted subcutaneously with an increasing number of HuT78 cells in the leg and displaced with an excess of cold antibody. We also studied irradiated severe combined immunodeficient (SCID) mice reconstituted with human peripheral blood mononuclear cells (hPBMCs) and injected with (99m)Tc-labeled visilizumab or control mAb. After dynamic imaging for 3 h, major organs were removed, counted, and processed for immunohistologic examination. RESULTS: Visilizumab was labeled with HYNIC with high labeling efficiency (>90%) and high specific activity (SA; 10,360-11,100 MBq/mg), with retained biochemical integrity and in vitro binding activity to CD3-positive cells. The in vivo targeting experiment showed a proportional increase of specific uptake with the number of injected cells, both at 6 and at 24 h, and the in vivo competition study demonstrated more than 60% decreased uptake after an excess of unlabeled antibody. In SCID mice, hPBMCs in different tissues were detected by (99m)Tc-labeled visilizumab and confirmed by histology. CONCLUSION: Visilizumab can be efficiently labeled with (99m)Tc with high efficiency and SA and could be a valuable tool for the study of human T-lymphocyte trafficking and lymphocytic infiltration of tissues and organs.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , CD3 Complex/immunology , Radioimmunodetection , T-Lymphocytes/metabolism , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Binding, Competitive , Cell Line , Cell Movement , Female , Humans , Immunoenzyme Techniques , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice , Molecular Probe Techniques , Niacinamide/analogs & derivatives , Niacinamide/chemistry , Organotechnetium Compounds/chemistry , Quality Control , Substrate Specificity , Succinimides/chemistry , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tissue Distribution
9.
Am J Respir Crit Care Med ; 178(10): 1002-8, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18787222

ABSTRACT

RATIONALE: Airway inflammation in asthma is associated with increased activated CD25(+) T cells, IL-2, and soluble IL-2 receptors (IL-2Rs). OBJECTIVES: A randomized, double-blinded, placebo-controlled study was used to evaluate the safety and efficacy of daclizumab, a humanized IgG1 monoclonal antibody against the IL-2R alpha chain (CD25) of activated lymphocytes, in adults with moderate to severe persistent asthma. METHODS: Patients with obstructive pulmonary functions, despite inhaled corticosteroids (ICS), were switched to equivalent dose inhaled triamcinolone acetate acetonide (TAA). Patients dependent on ICS were randomized (3:1) to daclizumab (intravenous loading dose, 2 mg/kg, then 1 mg/kg) or placebo every 2 weeks, added to stable-dose TAA through Week 12 (Treatment Period 1). Over Weeks 12-20 (Treatment Period 2), patients tapered TAA while on the study drug, and were followed for 16 weeks off the study drug. MEASUREMENTS AND MAIN RESULTS: Among 115 evaluable patients (88 daclizumab, 27 placebo), groups had similar age, disease duration, and length of ICS use. During Treatment Period 1, daclizumab improved FEV(1) (daclizumab, 4.4 +/- 1.80% vs. placebo, 1.5 +/- 2.39%; P = 0.05), and reduced daytime asthma symptoms (P = 0.018) and short-acting inhaled beta(2)-agonist use (P = 0.009). Daclizumab treatment prolonged time to exacerbation (P = 0.024). Adverse events were evenly distributed between groups, although there were more serious adverse events in the patients treated with daclizumab. CONCLUSIONS: Daclizumab improved pulmonary function and asthma control in patients with moderate to severe chronic asthma inadequately controlled on ICS. The mechanism of action likely involves inhibition of proinflammatory cytokine generation by IL-2R blockade in activated T cells. Clinical trial registered with www.clinicaltrials.gov (NCT00028288).


Subject(s)
Antibodies, Monoclonal/therapeutic use , Asthma/drug therapy , Immunoglobulin G/therapeutic use , Immunosuppressive Agents/therapeutic use , Interleukin-2/antagonists & inhibitors , Adult , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Asthma/physiopathology , Daclizumab , Double-Blind Method , Female , Forced Expiratory Volume/drug effects , Humans , Immunosuppressive Agents/pharmacokinetics , Interleukin-2 Receptor alpha Subunit , Kaplan-Meier Estimate , Male , Middle Aged , Pneumonia/drug therapy , Pneumonia/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
10.
Clin Cancer Res ; 14(9): 2775-84, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451245

ABSTRACT

PURPOSE: We generated a humanized antibody, HuLuc63, which specifically targets CS1 (CCND3 subset 1, CRACC, and SLAMF7), a cell surface glycoprotein not previously associated with multiple myeloma. To explore the therapeutic potential of HuLuc63 in multiple myeloma, we examined in detail the expression profile of CS1, the binding properties of HuLuc63 to normal and malignant cells, and the antimyeloma activity of HuLuc63 in preclinical models. EXPERIMENTAL DESIGN: CS1 was analyzed by gene expression profiling and immunohistochemistry of multiple myeloma samples and numerous normal tissues. HuLuc63-mediated antimyeloma activity was tested in vitro in antibody-dependent cellular cytotoxicity (ADCC) assays and in vivo using the human OPM2 xenograft model in mice. RESULTS: CS1 mRNA was expressed in >90% of 532 multiple myeloma cases, regardless of cytogenetic abnormalities. Anti-CS1 antibody staining of tissues showed strong staining of myeloma cells in all plasmacytomas and bone marrow biopsies. Flow cytometric analysis of patient samples using HuLuc63 showed specific staining of CD138+ myeloma cells, natural killer (NK), NK-like T cells, and CD8+ T cells, with no binding detected on hematopoietic CD34+ stem cells. HuLuc63 exhibited significant in vitro ADCC using primary myeloma cells as targets and both allogeneic and autologous NK cells as effectors. HuLuc63 exerted significant in vivo antitumor activity, which depended on efficient Fc-CD16 interaction as well as the presence of NK cells in the mice. CONCLUSIONS: These results suggest that HuLuc63 eliminates myeloma cells, at least in part, via NK-mediated ADCC and shows the therapeutic potential of targeting CS1 with HuLuc63 for the treatment of multiple myeloma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lymphocyte Subsets/metabolism , Multiple Myeloma/drug therapy , Plasma Cells/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Killer Cells, Natural/immunology , Lymphocyte Subsets/cytology , Mice , Mice, SCID , Multiple Myeloma/immunology , Plasma Cells/cytology , Receptors, Immunologic/genetics , Signaling Lymphocytic Activation Molecule Family , Xenograft Model Antitumor Assays
11.
Circ Res ; 91(10): 907-14, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12433835

ABSTRACT

Although inhibiting interaction of beta(2) integrins with cognate immunoglobulin class adhesion receptor ligands is an effective neuroprotective strategy in small mammal models of stroke, the strategy has failed in human trials. A completely different antiadhesion receptor strategy was therefore rigorously tested in a model that may more closely approximate human reperfused stroke. Early leukoadhesive events in postischemic cerebral microvessels are mediated by upregulated selectin-class adhesion receptors on endothelial cells. Therefore, a blocking antibody prepared against common P- and E-selectin epitopes was humanized to suppress complement activation and tested in a reperfused hemispheric stroke model in Papio anubis (baboon). Histological examination of postischemic cerebral microvessels revealed a strong upregulation of E-and P-selectin expression. Placebo-blinded administration of the humanized anti-human E- and P-selectin monoclonal antibody (HuEP5C7, 20 mg/kg IV, n=9; placebo, n=9) immediately after the onset of 1 hour of temporary ischemia resulted in trends showing reduced polymorphonuclear leukocyte (PMN) infiltration into ischemic cortex, reduced infarct volumes (by 41%), improved neurological score (by 35%), and improved ability to self-care (by 39%). Importantly, there was no evidence of systemic complement activation, immune suppression, or pathological coagulopathy associated with this therapy. These data suggest that a humanized anti-E/P-selectin antibody approach is safe and may be effective as a clinical treatment for human stroke.


Subject(s)
Antibodies, Monoclonal/therapeutic use , E-Selectin/immunology , Neuroprotective Agents/therapeutic use , P-Selectin/immunology , Stroke/therapy , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/genetics , Antibody Specificity/genetics , Antibody Specificity/immunology , Brain/blood supply , Brain/drug effects , Brain/pathology , Complement System Proteins/analysis , Complement System Proteins/drug effects , Disease Models, Animal , E-Selectin/blood , Humans , Intercellular Adhesion Molecule-1/drug effects , Intercellular Adhesion Molecule-1/immunology , Leukocyte Count , Magnetic Resonance Angiography , Neuroprotective Agents/adverse effects , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , P-Selectin/blood , Papio , Prospective Studies , Single-Blind Method , Species Specificity , Stroke/blood , Stroke/immunology , Stroke/pathology , Treatment Outcome , Vascular Cell Adhesion Molecule-1/drug effects , Vascular Cell Adhesion Molecule-1/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...