Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Curr Pharm Biotechnol ; 14(2): 242-8, 2013.
Article En | MEDLINE | ID: mdl-23157711

CNTO 530 is an erythropoietin receptor agonist MIMETIBODYTM construct. CNTO 530 has been shown to be active in a number of rodent models of acquired anemia (e.g. renal insufficiency and chemotherapy induced anemia). We investigated the efficacy of CNTO 530 in murine models of ß-thalassemia and sickle cell anemia (Berkeley mice). ß- thalassemic mice are deficient in expression of α-globin chain and heterozygous mice are characterized by a clinical syndrome similar to the human ß-thalassemia intermedia. Berkeley mice are knocked out for murine alpha and beta globin and are transgenic for human alpha, beta (sickle) and gamma globin genes. Berkeley mice thus express human sickle hemoglobin A (HbS) and can also express human fetal hemoglobin. These mice express a severe compensated hypochromic microcytic anemia and display the sickle cell phenotype. To test the effectiveness of CNTO 530, mice from both genotypes received a single subcutaneous (s.c.) dose of CNTO 530 or darbepoetin-α (as a comparator) at 10,000 U/kg, a dose shown to cause a similar increase in reticulocytes and hemoglobin in normal mice. Hematologic parameters were evaluated over time. CNTO 530, but not darbepoetin-α, increased reticulocytes, red blood cells and total hemoglobin in ß- thalassemic mice. In Berkeley mice CNTO 530 showed an increase in reticulocytes, red blood cells, F-cells, total hemoglobin and fetal hemoglobin. In conclusion, CNTO 530 is effective in murine models of ß-thalassemia and sickle cell anemia. These data suggest that CNTO 530 may have beneficial effects in patients with genetically mediated hemoglobinopathies.


Anemia, Sickle Cell/drug therapy , Receptors, Erythropoietin/agonists , Recombinant Fusion Proteins/therapeutic use , beta-Thalassemia/drug therapy , Anemia, Sickle Cell/blood , Animals , Darbepoetin alfa , Disease Models, Animal , Erythrocyte Count , Erythropoietin/analogs & derivatives , Erythropoietin/pharmacology , Erythropoietin/therapeutic use , Female , Hematinics/pharmacology , Hematinics/therapeutic use , Hemoglobins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recombinant Fusion Proteins/pharmacology , beta-Thalassemia/blood
2.
J Immunotoxicol ; 9(1): 43-55, 2012.
Article En | MEDLINE | ID: mdl-22299716

Many immunosuppressive drugs are associated with an increased risk of neoplasia, principally non-melanoma skin cancers and B-cell lymphomas. However, only 6 of the 13 immunosuppressive drugs tested in 2 year bioassays increased the incidence of neoplasia. For example, the 2-year bioassays conducted with cyclosporine (CSA), an International Agency for Research on Cancer (IARC) Group 1 human carcinogen, were negative. The purpose of these investigations was to use transplanted tumor models in immunocompetent, syngeneic mice to gain insight into the failure of the 2-year bioassay to show an increased incidence of neoplasia with CSA. C3H HeN mice were used in a battery of assays with a transplanted squamous cell carcinoma (SCC VII cells) or a B-cell, lymphoma (38C13 cells) cells to study effects of CSA on local growth and metastases, experimental metastases, and progression of established metastases. Mice received CSA twice weekly by subcutaneous (SC) injection at doses of 0.5, 5, or 50 mg/kg; controls received the CSA vehicle. CSA had a modest inhibitory effect on SC tumors initiated by 38C13 cells and on intramuscular tumors initiated by SCC VII cells. CSA also decreased the number of lung colonies and decreased the size, growth fraction and vascularity of established lung metastases initiated by SCC VII cells. In contrast, CSA increased progressive growth of metastases to the sentinel lymph node from an intramuscular SCC VII tumor, but had no effect cellular traffic to the node. In conclusion, CSA at doses up to 50 mg/kg did not facilitate tumor progression and it partially inhibited tumor growth, suggesting that suppression of tumor progression may partially explain the failure of CSA to act as a carcinogen in 2 year bioassays.


Carcinoma, Squamous Cell/secondary , Cyclosporine/toxicity , Immunosuppressive Agents/toxicity , Lung Neoplasms/secondary , Lymphoma, B-Cell/pathology , Muscle Neoplasms/pathology , Animals , Carcinogenicity Tests , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/prevention & control , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclosporine/administration & dosage , Disease Progression , Dose-Response Relationship, Drug , Female , Immunosuppressive Agents/administration & dosage , Injections, Subcutaneous , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lymphatic Metastasis , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/prevention & control , Mice , Mice, Inbred C3H , Muscle Neoplasms/immunology , Muscle Neoplasms/prevention & control , Neoplasm Invasiveness , Risk Assessment , Time Factors , Tumor Burden/drug effects
3.
Br J Pharmacol ; 166(3): 823-46, 2012 Jun.
Article En | MEDLINE | ID: mdl-22168282

Monoclonal antibodies (mAbs) and fusion proteins directed towards cell surface targets make an important contribution to the treatment of disease. The purpose of this review was to correlate the clinical and preclinical data on the 15 currently approved mAbs and fusion proteins targeted to the cell surface. The principal sources used to gather data were: the peer reviewed Literature; European Medicines Agency 'Scientific Discussions'; and the US Food and Drug Administration 'Pharmacology/Toxicology Reviews' and package inserts (United States Prescribing Information). Data on the 15 approved biopharmaceuticals were included: abatacept; abciximab; alefacept; alemtuzumab; basiliximab; cetuximab; daclizumab; efalizumab; ipilimumab; muromonab; natalizumab; panitumumab; rituximab; tocilizumab; and trastuzumab. For statistical analysis of concordance, data from these 15 were combined with data on the approved mAbs and fusion proteins directed towards soluble targets. Good concordance with human pharmacodynamics was found for mice receiving surrogates or non-human primates (NHPs) receiving the human pharmaceutical. In contrast, there was poor concordance for human pharmacodynamics in genetically deficient mice and for human adverse effects in all three test systems. No evidence that NHPs have superior predictive value was found.


Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/toxicity , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/toxicity , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Drug Evaluation, Preclinical , Humans , Maximum Tolerated Dose , No-Observed-Adverse-Effect Level , Pharmacology, Clinical , Predictive Value of Tests , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Solubility , Species Specificity
4.
Br J Pharmacol ; 166(3): 806-22, 2012 Jun.
Article En | MEDLINE | ID: mdl-22168335

Monoclonal antibodies (mAbs) and fusion proteins directed towards soluble targets make an important contribution to the treatment of disease. The purpose of this review was to correlate the clinical and preclinical data on the 14 currently approved mAbs and fusion proteins targeted to soluble targets. The principal sources used to gather data were: the peer reviewed Literature; European Medicines Agency 'Scientific Discussions' and United States Food and Drug Administration 'Pharmacology/Toxicology Reviews' and package inserts (United States Prescribing Information). Data on the following approved biopharmaceuticals were included: adalimumab, anakinra, bevacizumab, canakinumab, certolizumab pegol, denosumab, eculizumab, etanercept, golimumab, infliximab, omalizumab, ranibizumab, rilonacept and ustekinumab. Some related biopharmaceuticals in late-stage development were also included for comparison. Good concordance with human pharmacodynamics was found for both non-human primates (NHPs) receiving the human biopharmaceutical and mice receiving rodent homologues (surrogates). In contrast, there was limited concordance for human adverse effects in genetically deficient mice, mice receiving surrogates or NHPs receiving the human pharmaceutical. In summary, the results of this survey show that although both mice and NHPs have good predictive value for human pharmacodynamics, neither species have good predictive value for human adverse effects. No evidence that NHPs have superior predictive value was found.


Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/toxicity , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/toxicity , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Drug Evaluation, Preclinical , Humans , Maximum Tolerated Dose , No-Observed-Adverse-Effect Level , Pharmacology, Clinical , Predictive Value of Tests , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Solubility , Species Specificity
5.
Exp Diabetes Res ; 2011: 910159, 2011.
Article En | MEDLINE | ID: mdl-21754921

Patients treated with recombinant human Epo demonstrate an improvement in insulin sensitivity. We aimed to investigate whether CNTO 530, a novel Epo receptor agonist, could affect glucose tolerance and insulin sensitivity. A single administration of CNTO 530 significantly and dose-dependently reduced the area under the curve in a glucose tolerance test in diet-induced obese and diabetic mice after 14, 21, and 28 days. HOMA analysis suggested an improvement in insulin sensitivity, and this effect was confirmed by a hyperinsulinemic-euglycemic clamp. Uptake of (14)C-2-deoxy-D-glucose indicated that animals dosed with CNTO 530 transported more glucose into skeletal muscle and heart relative to control animals. In conclusion, CNTO530 has a profound effect on glucose tolerance in insulin-resistant rodents likely because of improving peripheral insulin sensitivity. This effect was observed with epoetin-α and darbepoetin-α, suggesting this is a class effect, but the effect with these compounds relative to CNTO530 was decreased in duration and magnitude.


Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Receptors, Erythropoietin/agonists , Recombinant Fusion Proteins/pharmacology , Animals , Darbepoetin alfa , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/physiopathology , Dietary Fats/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Epoetin Alfa , Erythropoietin/analogs & derivatives , Erythropoietin/pharmacology , Glucose Clamp Technique , Glucose Tolerance Test , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/complications , Obesity/etiology , Obesity/metabolism , Recombinant Proteins , Time Factors
6.
Int Immunopharmacol ; 11(11): 1697-705, 2011 Nov.
Article En | MEDLINE | ID: mdl-21689786

Anti-CD28 superagonist (SA) mediated cytokine release syndrome (CRS), an adverse event resulting in systemic release of cytokines, is an emergent issue in drug development. CRS is of potential concern for all monoclonal antibodies (mAbs) particularly those directed against cell surface targets on lymphocytes. Concern regarding patient safety requires development of novel methods to predict these adverse reactions. Due to the inability of animal studies to predict CRS, we have developed a whole blood in vitro screen to support First in Human studies and assess the potential for mAbs to cause anti-CD28 SA-like CRS. For this purpose we have immobilized marketed mAbs, whose potential for causing CRS and milder infusion reactions is known, on Protein A beads and used these beads to stimulate cytokine release. After culture, supernatants are harvested and frozen for later multiplex analysis of cytokines using Searchlight™ technology. We have employed hierarchicalluster analysis (HCA) to allow comparison of 12 different cytokine levels across numerous donors, treatments, and experiments. Results conclusively distinguish test mAb responses from an anti-CD28 superagonist mAb response. As part of a global analysis of preclinical data, the results of this assay can facilitate entry into First in Human clinical trials, help with selection of starting doses and may allow more rapid dose escalation using smaller cohorts.


Antibodies, Monoclonal/adverse effects , CD28 Antigens/immunology , Cytokines/blood , Drug-Related Side Effects and Adverse Reactions/blood , Enzyme-Linked Immunosorbent Assay/methods , Immune System Diseases/blood , Cluster Analysis , Cytokines/immunology , Drug-Related Side Effects and Adverse Reactions/immunology , Drug-Related Side Effects and Adverse Reactions/metabolism , Enzyme-Linked Immunosorbent Assay/statistics & numerical data , Humans , Immune System Diseases/chemically induced , Immune System Diseases/metabolism , Predictive Value of Tests , Receptors, Fc/metabolism , Syndrome
7.
J Pharmacol Toxicol Methods ; 63(3): 236-49, 2011.
Article En | MEDLINE | ID: mdl-21156211

INTRODUCTION: Immunosuppressive drugs are associated with an increased risk of infections and in some cases neoplasia, particularly non-melanoma skin cancers. This paper describes the development of a model to test the effects of immunosuppressive drugs on local invasion and metastases of a squamous cell carcinoma in syngeneic, immunocompetent mice. METHODS: SCC VII cells were labeled with 655 quantum dots (QDs), injected intramuscularly into C3H HEN mice and traffic and progressive growth in the draining popliteal lymph node were evaluated. RESULTS: SCC VII cells express RAE-1, an NKG2D ligand, and were sensitive to natural killer (NK) cells in vitro. QDs were stable in SCC VII cells and showed no evidence of toxicity to the cells. In vivo, confocal microscopy showed that QD-labeled SCC VII cells could migrate to the draining node and microfluorimetry showed progressive traffic of QDs to the node. There was no evidence of systemic toxicity of QDs. Primary immunosuppression in SCID and SCID-beige mice and treatment of normal mice with immunosuppressive agents (anti-asialoGM1 and cyclophosphamide) can enhance traffic of QDs and/or metastases to the draining lymph node. In contrast, cyclosporine had no effect on traffic or metastases. CONCLUSION: This model of local invasion and metastases may be useful in immunotoxicology for identifying and characterizing the hazard posed by selective immunosuppressive drugs.


Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/secondary , Immunosuppressive Agents/toxicity , Neoplasms, Experimental/immunology , Neoplasms, Experimental/secondary , Animals , Cell Culture Techniques , Cell Line, Tumor , Flow Cytometry , Immunohistochemistry , Lymphatic Metastasis , Mice , Mice, Inbred C3H , Mice, SCID , Neoplasm Transplantation
8.
Int J Toxicol ; 29(5): 435-66, 2010.
Article En | MEDLINE | ID: mdl-20884856

Many immunosuppressive drugs are associated with an increased risk of B-cell lymphoma, squamous cell carcinoma, and Kaposi sarcoma. Thirteen immunosuppressive drugs have been tested in 2-year carcinogenicity studies (abatacept; azathioprine; busulfan; cyclophosphamide; cyclosporine; dexamethasone; everolimus; leflunomide; methotrexate; mycophenolate mofetil; prednisone; sirolimus; and tacrolimus) and in additional models including neonatal and genetically modified mice; chemical, viral, ultraviolet, and ionizing radiation co-carcinogenesis, and in models with transplanted tumor cells. The purpose of this review is to outline the mechanisms by which immunosuppressive drugs can influence neoplasia, to summarize the available preclinical data on the 13 drugs, and to critically review the performance of the models. A combination of primary tumor and metastasis assays conducted with transplanted cells may provide the highest value for hazard identification and can be applied on a case-by-case basis. However, for both small molecules and therapeutic proteins, determining the relative risk to patients from preclinical data remains problematic. Classifying immunosuppressive drugs based on their mechanism of action and hazard identification from preclinical studies and a prospective pharmacovigilance program to monitor carcinogenic risk may be a feasible way to manage patient safety during the clinical development program and postmarketing.


Immunologic Surveillance/drug effects , Immunosuppression Therapy/adverse effects , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacology , Neoplasms/epidemiology , Animals , Drug Evaluation, Preclinical/methods , Humans , Immunologic Surveillance/physiology , Immunosuppressive Agents/classification , Neoplasms/physiopathology , Organ Transplantation , Product Surveillance, Postmarketing , Toxicity Tests, Chronic/methods
9.
MAbs ; 2(4): 428-39, 2010.
Article En | MEDLINE | ID: mdl-20519961

We prepared and characterized golimumab (CNTO148), a human IgG1 tumor necrosis factor alpha (TNFα) antagonist monoclonal antibody chosen for clinical development based on its molecular properties. Golimumab was compared with infliximab, adalimumab and etanercept for affinity and in vitro TNFα neutralization. The affinity of golimumab for soluble human TNFα, as determined by surface plasmon resonance, was similar to that of etanercept (18 pM versus 11 pM), greater than that of infliximab (44 pM) and significantly greater than that of adalimumab (127 pM, p=0.018).  The concentration of golimumab necessary to neutralize TNFα-induced E-selectin expression on human endothelial cells by 50% was significantly less than those for infliximab (3.2 fold; p=0.017) and adalimumab (3.3-fold; p=0.008) and comparable to that for etanercept. The conformational stability of golimumab was greater than that of infliximab (primary melting temperature [Tm] 74.8 °C vs. 69.5 °C) as assessed by differential scanning calorimetry.  In addition, golimumab showed minimal aggregation over the intended shelf life when formulated as a high concentration liquid product (100 mg/mL) for subcutaneous administration.  In vivo, golimumab at doses of 1 and 10 mg/kg significantly delayed disease progression in a mouse model of human TNFα-induced arthritis when compared with untreated mice, while infliximab was effective only at 10 mg/kg. Golimumab also significantly reduced histological scores for arthritis severity and cartilage damage, as well as serum levels of pro-inflammatory cytokines and chemokines associated with arthritis. Thus, we have demonstrated that golimumab is a highly stable human monoclonal antibody with high affinity and capacity to neutralize human TNFα in vitro and in vivo.


Antibodies, Monoclonal/pharmacology , Arthritis/immunology , Cartilage/drug effects , Immunoglobulin G/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Adalimumab , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Affinity , Arthritis/chemically induced , Cartilage/pathology , Disease Models, Animal , Disease Progression , E-Selectin/genetics , E-Selectin/metabolism , Etanercept , Gene Expression Regulation/drug effects , Hybridomas , Immunoglobulin G/isolation & purification , Inflammation Mediators/metabolism , Infliximab , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Protein Conformation , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/immunology
10.
Article En | MEDLINE | ID: mdl-20077575

BACKGROUND: CNTO 530is a biopharmaceutical consisting of a novel peptide that mimics the actions of erythropoietin, fused to the Fc fragment of human IgG4. Pharmacokinetic and pharmacodynamic studies showed that CNTO 530 produced sustained increases in red blood cell parameters in rats and rabbits and that the serum half life of CNTO 530 was 2 days in rabbits and 3 days in rats. METHODS: For the evaluation of embryofetal development, CNTO 530 was injected at loading doses of 0, 0.9/1, 6, or 60 mg/kg subcutaneously (SC) on gestation day (GD)7 followed by maintenance doses of 0, 0.3, 2, or 20 mg/kg SC every 3 days through GD16 in rats and every 2 days through GD19 in rabbits (GD0 was the day of mating). Rats were Caesarean sectioned on GD21, rabbits on GD29. RESULTS: Administration of CNTO 530 was associated with an increase in hematocrit at all dose levels and a decrease in maternal body weight gains. Fetuses exhibited reduced body weight and delayed ossification. Soft tissue changes were limited to cardiovascular alterations in the high-dose rabbits only. Rat and rabbit fetuses were exposed to CNTO 530 in all dose groups. CONCLUSIONS: These studies show that the embryo/fetal development effects observed following CNTO 530 treatment during organogenesis are qualitatively similar to those seen with other erythropoietin agonists and are likely a secondary consequence of increased hematocrit in the dams. Unlike other erythropoietin receptor agonists, CNTO 530 was able to cross the placental barrier, which was considered likely the result of FcRn-mediated transcytosis.


Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Fetal Development/drug effects , Receptors, Erythropoietin/agonists , Recombinant Fusion Proteins/toxicity , Teratogens/toxicity , Abnormalities, Drug-Induced/etiology , Animals , Body Weight/drug effects , Bone and Bones/drug effects , Bone and Bones/embryology , Embryo, Mammalian/embryology , Female , Fetal Weight/drug effects , Injections, Subcutaneous , Osteogenesis/drug effects , Polycythemia/chemically induced , Pregnancy , Rabbits , Rats , Recombinant Fusion Proteins/classification , Teratogens/classification
11.
Curr Opin Drug Discov Devel ; 13(1): 124-35, 2010 Jan.
Article En | MEDLINE | ID: mdl-20047153

Infusion reactions and cytokine release syndrome (CRS) are an emerging issue in drug development and are of particular importance with the development of new therapeutic proteins. Increasing concerns regarding patient safety require a better understanding of the mechanism involved and the development of novel methods for preventing and predicting such reactions and CRS. This review discusses developments during the past few years in understanding the mechanisms that cause infusion reactions and CRS, advances in approaches to prevent CRS, the reason why preclinical animal models are unreliable predictors of CRS, and new developments in the design and analysis of in vitro screening systems for the prediction of CRS.


Cytokines/metabolism , Drug-Related Side Effects and Adverse Reactions/chemically induced , Drug-Related Side Effects and Adverse Reactions/immunology , Immune System Diseases/chemically induced , Immune System Diseases/metabolism , Animals , Cluster Analysis , Cytokines/immunology , Humans , Receptors, Fc/metabolism
12.
Respir Res ; 10: 43, 2009 Jun 01.
Article En | MEDLINE | ID: mdl-19486528

BACKGROUND: The immune mechanisms associated with infection-induced disease exacerbations in asthma and COPD are not fully understood. Toll-like receptor (TLR) 3 has an important role in recognition of double-stranded viral RNA, which leads to the production of various inflammatory mediators. Thus, an understanding of TLR3 activation should provide insight into the mechanisms underlying virus-induced exacerbations of pulmonary diseases. METHODS: TLR3 knock-out (KO) mice and C57B6 (WT) mice were intranasally administered repeated doses of the synthetic double stranded RNA analog poly(I:C). RESULTS: There was a significant increase in total cells, especially neutrophils, in BALF samples from poly(I:C)-treated mice. In addition, IL-6, CXCL10, JE, KC, mGCSF, CCL3, CCL5, and TNFalpha were up regulated. Histological analyses of the lungs revealed a cellular infiltrate in the interstitium and epithelial cell hypertrophy in small bronchioles. Associated with the pro-inflammatory effects of poly(I:C), the mice exhibited significant impairment of lung function both at baseline and in response to methacholine challenge as measured by whole body plethysmography and an invasive measure of airway resistance. Importantly, TLR3 KO mice were protected from poly(I:C)-induced changes in lung function at baseline, which correlated with milder inflammation in the lung, and significantly reduced epithelial cell hypertrophy. CONCLUSION: These findings demonstrate that TLR3 activation by poly(I:C) modulates the local inflammatory response in the lung and suggest a critical role of TLR3 activation in driving lung function impairment. Thus, TLR3 activation may be one mechanism through which viral infections contribute toward exacerbation of respiratory disease.


Inflammation/chemically induced , Poly I-C/pharmacology , Toll-Like Receptor 3/physiology , Animals , Cell Line , Cytokines/metabolism , Female , Humans , Inflammation/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plethysmography , Respiratory Function Tests , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/genetics
13.
Blood ; 113(20): 4955-62, 2009 May 14.
Article En | MEDLINE | ID: mdl-19264917

Anemia as associated with numerous clinical conditions can be debilitating, but frequently can be treated via administration of epoetin-alfa, darbepoietin-alfa, or methoxy-PEG epoetin-beta. Despite the complexity of EPO-EPO receptor interactions, the development of interesting EPO mimetic peptides (EMPs) also has been possible. CNTO 530 is one such novel MIMETIBODY Fc-domain dimeric EMP fusion protein. In a mouse model, single-dose CNTO 530 (unlike epoetin-alfa or darbepoietin-alfa) bolstered red cell production for up to 1 month. In 5-fluorouracil and carboplatin-paclitaxel models, CNTO 530 also protected against anemia with unique efficiency. These actions were not fully accounted for by half-life estimates, and CNTO 530 signaling events therefore were studied. Within primary bone marrow erythroblasts, kinetics of STAT5, ERK, and AKT activation were similar for CNTO 530 and epoetin-alfa. p70S6K activation by CNTO 530, however, was selectively sustained. In vivo, CNTO 530 uniquely stimulated the enhanced formation of PODXL(high)CD71(high) (pro)erythroblasts at frequencies multifold above epoetin-alfa or darbepoietin-alfa. CNTO 530 moreover supported the sustained expansion of a bone marrow-resident Kit(neg)CD71(high)Ter119(neg) progenitor pool. Based on these distinct erythropoietic and EPOR signaling properties, CNTO 530 holds excellent promise as a new EPO mimetic.


Bone Marrow Cells/drug effects , Erythroblasts/drug effects , Erythropoietin/analogs & derivatives , Hematinics/pharmacology , Recombinant Fusion Proteins/pharmacology , Anemia/pathology , Animals , Bone Marrow Cells/physiology , Cell Count , Cells, Cultured , Drug Evaluation, Preclinical , Erythroblasts/physiology , Erythropoiesis/drug effects , Erythropoietin/chemistry , Female , Hematinics/chemistry , Mice , Mice, Inbred C57BL , Molecular Mimicry , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/chemistry , Signal Transduction/drug effects , Time Factors
14.
Expert Rev Clin Immunol ; 5(5): 499-521, 2009 Sep.
Article En | MEDLINE | ID: mdl-20477639

Monoclonal antibodies (mAbs) are widely used in anti-inflammatory and tumor therapy. Although effective, mAbs can cause a variety of adverse effects. An important toxicity seen with a few mAbs is cytokine-release syndrome (CRS). These mAbs include: alemtuzumab, muromonab-CD3, rituximab, tosituzumab, CP-870,893, LO-CD2a/BTI-322 and TGN1412. By contrast, over 30 mAbs used clinically are not associated with CRS. In this review, the clinical aspects of CRS, the mAbs associated with CRS, the cytokines involved and putative mechanisms mediating cytokine release will be discussed. This will be followed by a discussion of the poor predictive value of studies in animals and the prospects for creating in vitro screens. Finally, approaches to decreasing the probability of CRS, decreasing the severity or treating CRS, should it occur, will be described.

15.
Cytometry A ; 73(8): 702-14, 2008 Aug.
Article En | MEDLINE | ID: mdl-18496852

Analysis of multicolor flow cytometric data is traditionally based on the judgment of an expert, generally time consuming, sometimes incomplete and often subjective in nature. In this article, we investigate another statistical method using a Sequential Univariate Gating (SUG) algorithm to identify regions of interest between two groups of multivariate flow cytometric data. The metric used to differentiate between the groups of univariate distributions in SUG is the Kolmogorov-Smirnov distance (D) statistic. The performance of the algorithm is evaluated by applying it to a known three-color data set looking at activation of CD4+ and CD8+ lymphocytes with anti-CD3 antibody treatment and comparing the results to the expert analysis. The algorithm is then applied to a four-color data set used to study the effects of recombinant human erythropoietin (rHuEPO) on several murine bone marrow populations. SUG was used to identify regions of interest in the data and results compared to expert analysis and the current state-of-the-art statistical method, Frequency Difference Gating (FDG). Cluster analysis was then performed to identify subpopulations responding differently to rHuEPO. Expert analysis, SUG and FDG identified regions in the data that showed activation of CD4+ and CD8+ lymphocytes with anti-CD3 treatment. In the rHuEPO treated data sets, the expert and SUG identified a dose responsive expansion of only the erythroid precursor population. In contrast, FDG resulted in identification of regions of interest both in the erythroid precursors as well as in other bone marrow populations. Clustering within the regions of interest defined by SUG resulted in identification of four subpopulations of erythroid precursors that are morphologically distinct and show a differential response to rHuEPO treatment. Greatest expansion is seen in the basophilic and poly/orthochromic erythroblast populations with treatment. Identification of populations of interest can be performed using SUG in less subjective, time efficient, biologically interpretable manner that corroborates with the expert analysis. The results suggest that basophilic erythroblasts cells or their immediate precursors are an important target for the effects of rHuEPO in murine bone marrow. The MATLAB implementation of the method described in the article, both experimental data and other supplemental materials are freely available at http://web.mac.com/acidrap18.


Algorithms , Bone Marrow Cells/drug effects , Erythropoietin/pharmacology , Flow Cytometry/methods , Animals , Antibodies/pharmacology , CD3 Complex , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Erythroid Cells/cytology , Erythroid Cells/drug effects , Humans , Mice , Mice, Inbred C57BL , Recombinant Proteins , Spleen/drug effects , Stem Cells/cytology , Stem Cells/drug effects
16.
Cytometry A ; 73(2): 148-59, 2008 Feb.
Article En | MEDLINE | ID: mdl-18205195

TNF-alpha is a pleitropic cytokine that expresses both pro- and anti-inflammatory activity and transgenic mice expressing human tumor necrosis factor-alpha (TNF-alpha) exhibit a progressive polyarthritis that models rheumatoid arthritis (RA). One of the common comorbidities of RA is anemia of chronic disease (ACD). The purpose of these experiments was to study the changes in the bone marrow and peripheral blood that accompany polyarthritis in TNF-alpha transgenic mice in an effort to better understand the pathogenesis of myelodysplasia and ACD. Polychromatic cytometry, hematology and serum cytokine analysis were used to study the pathogenesis of ACD in human TNF-alpha transgenic mice. Our hematological evaluation revealed a mild, compensated, microcytic hypochromic anemia, and monocytosis. In the bone marrow, we observed alterations in cell kinetics, decreased relative expression of transferrin receptor and increased apoptosis and cell death in several late precursor cell populations. Although significant levels of human TNF-alpha were found in the serum, neither change in serum murine erythropoietin nor any significant difference observed in serum levels of murine IL-beta, IL-5, IL-6, IL-10, IL-12(p70), IL-17, TNF-alpha, IFNgamma, GM-CSF, MIP-1alphaJE, MCP-5 was observed. Tg197 mice develop a compensated, microcytic, hypochromic anemia, and a functional iron deficiency by 9 weeks of age. Changes in peripheral blood are reflected in alterations in cell kinetics, transferrin receptor expression and markedly increased apoptosis and cell death in the bone marrow indicating that TNF-alpha may contribute to myelodysplasia in ACD. Moreover, since human TNF-alpha can interact only with murine TNFR1, our data suggest that TNFR1 may play an important role in the development of ACD.


Anemia, Hypochromic/pathology , Arthritis/pathology , Cytokines/blood , Myelodysplastic Syndromes/pathology , Tumor Necrosis Factor-alpha/physiology , Anemia, Hypochromic/metabolism , Animals , Apoptosis/physiology , Arthritis/metabolism , Bone Marrow/metabolism , Cell Death/physiology , Chronic Disease , Humans , Joint Capsule/metabolism , Joint Capsule/pathology , Mice , Mice, Transgenic , Myelodysplastic Syndromes/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/genetics
17.
Pharm Res ; 25(2): 369-78, 2008 Feb.
Article En | MEDLINE | ID: mdl-17609863

PURPOSE: Originally approved for three times/week dosing, recombinant human erythropoietin (rhEPO) is now often used at weekly intervals. We have studied rhEPO in mice to better understand why the extended dosing interval retains efficacy. METHODS: C57Bl/6 mice received a single sc. dose of rhEPO (3,000 IU/kg). Bone marrow and blood were collected at 8 h and 1, 2, 5 and 7 days. Staining for TER-119 and CD71, pulse labeling with bromodeoxyuridine, annexin-V binding and vital staining with 7-aminoactinomycin D: were used cell cycle and apoptosis in erythroblasts by four color flow cytometry. RESULTS: A wave of proliferation and/or maturation progressed through all erythroblasts, resulting in the emigration of immature reticulocytes into the periphery. An increase in the fraction of erythroblasts in S and G2M was found, but suppression of apoptosis was not. CONCLUSIONS: Most of the effects of rhEPO occurred 48 h after dosing, when the concentration of rhEPO was less than 1% of Cmax, suggesting that the processes set in motion by rhEPO can continue after rhEPO concentrations fall. Our observation of apoptosis in erythroblasts even when rhEPO concentrations were high suggests that regulatory mechanisms which down-regulate erythropoiesis are also engaged.


Bone Marrow/drug effects , Erythropoietin/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Dose-Response Relationship, Drug , Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacokinetics , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Recombinant Proteins , Reference Values , Reticulocytes/drug effects
18.
Inflamm Allergy Drug Targets ; 6(3): 142-9, 2007 Sep.
Article En | MEDLINE | ID: mdl-17897050

Classically, B-lymphocytes (B cells) are considered to be the mediators of humoral immunity and their role in inflammatory disease largely confined to the down-stream function of antigen-antibody complexes, e.g., in fixing complement and mediating antibody dependent cellular cytotoxicity. More recently, and with the growing acceptance of the view that the immune system operates as an interconnected web of cells and cytokines, a larger role for B cells has been proposed. In this review, with a focus on how B cells and their cytokine products may present novel therapeutic targets, we will briefly discuss B cell ontogeny and discuss the evidence supporting a larger role for B cells in a variety of inflammatory diseases. Special emphasis will be placed on autoimmune diseases. These discussions are intended to provide the reader with the basis for viewing B cells as players of a broader role in inflammatory disease and thus suggest avenues for exploiting B cell directed therapy in novel ways.


B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , B-Lymphocytes/drug effects , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/prevention & control , Models, Immunological
19.
Transplantation ; 84(3): 308-15, 2007 Aug 15.
Article En | MEDLINE | ID: mdl-17700154

BACKGROUND: Tissue factor (TF) expression on islets can result in an instant blood-mediated inflammatory reaction (IBMIR) that contributes to early islet loss. We tested whether peritransplant protection of islets from IBMIR with a monoclonal anti-TF antibody (CNTO859) would enhance engraftment in our nonhuman primate marginal mass model. METHODS: Each of six pairs of cynomolgus monkeys (CM) with streptozotocin-induced diabetes was closely matched for metabolic control and was transplanted with 5,000 IEQ/kg allogeneic, ABO-compatible islets from the same donor under the cover of steroid-free immunosuppression. For each pair, experimental animals received islets cultured with 20 microg/mL anti-TF and were dosed with 6 mg/kg anti-TF intravenously, 10-25 min before islet infusion; control monkeys received an equal number of islets from the same preparation cultured without anti-TF and no in vivo treatment. RESULTS: Early fasting C-peptide (CP) values were different between (P<0.01), but not within, pairs and correlated with in vitro functional capacity of islets as assessed by perifusion (r=0.60; P=0.022). Compared to their matched controls, experimental animals had decreased posttransplant markers of coagulation, higher fasting CP levels (1 month posttransplant and end of study) and prolonged graft function. CONCLUSIONS: These data suggest that pretreatment of islets and the recipient with anti-TF may limit the effects of IBMIR, thereby enhancing islet engraftment and survival.


Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Thromboplastin/physiology , Transplantation Tolerance/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Survival/drug effects , Diabetes Mellitus, Experimental/surgery , Dose-Response Relationship, Drug , Fibrinolysis/immunology , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Macaca fascicularis , Models, Biological , Streptozocin , Thromboplastin/drug effects
20.
Cytometry A ; 71(8): 612-24, 2007 Aug.
Article En | MEDLINE | ID: mdl-17542025

BACKGROUND: Cellular binding of annexin V and membrane permeability to 7-aminoactinomycin D (7AAD) are important tools for studying apoptosis and cell death by flow cytometry. Combining viability markers with cell surface marker expression is routinely used to study various cell lineages. Current classification methods using strict thresholds, or "gates," on the fluorescent intensity of these markers are subjective in nature and may not fully describe the phenotypes of interest. We have developed objective criteria for phenotypic boundary recognition through the application of statistical pattern recognition. This task was achieved using artificial neural networks (ANNs) that were trained to recognize subsets of cells with known phenotypes, and then used to determine decision boundaries based on statistical measures of similarity. This approach was then used to test the hypothesis that erythropoietin (EPO) inhibits apoptosis and cell death in erythroid precursor cells in murine bone marrow. METHODS: Our method was developed for classification of viability using an in vitro cell system and then applied to an ex vivo analysis of murine late-stage erythroid progenitors. To induce apoptosis and cell death in vitro, an EPO-dependent human leukemic cell line, UT-7(EPO) cells were incubated without recombinant human erythropoietin (rhEPO) for 72 h. Five different ANNs were trained to recognize live, apoptotic, and dead cells using a "known" subset of the data for training, and a K-fold cross validation procedure for error estimation. The ANNs developed with the in vitro system were then applied to classify cells from an ex vivo study of rhEPO treated mice. Tg197 (human tumor necrosis-alpha transgenic mice, a model of anemia of chronic disease) received a single s.c. dose of 10,000 U/kg rhEPO and femoral bone marrow was collected 1, 2, 4, and 8 days after dosing. Femoral bone marrow cells were stained with TER-119 PE, CD71 APC enable identification of erythroid precursors, and annexin V FITC and 7AAD to identify the apoptotic and dead cells. During classification forward and side angle light scatter were also input to all pattern recognition systems. RESULTS: Similar decision boundaries between live, apoptotic, and dead cells were consistently identified by the neural networks. The best performing network was a radial basis function multi-perceptron that produced an estimated average error rate of 4.5% +/- 0.9%. Using these boundaries, the following results were reached: depriving UT-7(EPO) cells of rhEPO induced apoptosis and cell death while the addition of rhEPO rescued the cells in a dose-dependent manner. In vivo, treatment with rhEPO resulted in an increase of live erythroid cells in the bone marrow to 119.8% +/- 9.8% of control at the 8 day time point. However, a statistically significant transient increase in TER-119(+) CD71(+) 7AAD(+) dead erythroid precursors was observed at the 1 and 2 day time points with a corresponding decrease in TER-119(+) CD71(+) 7AAD(-) Annexin V(-) live erythroid precursors, and no change in the number of TER-119(+) CD71(+) annexin V(+) 7AAD(-) apoptotic erythroid precursors in the bone marrow. CONCLUSIONS: A statistical pattern recognition approach to viability classification provides an objective rationale for setting decision boundaries between "positive" and "negative" intensity measures in cytometric data. Using this approach we have confirmed that rhEPO inhibits apoptosis and cell death in an EPO dependent cell line in vitro, but failed to do so in vivo, suggesting EPO may not act as a simple antiapoptotic agent in the bone marrow. Rather, homeostatic mechanisms may regulate the pharmacodynamic response to rhEPO.


Bone Marrow Cells/cytology , Cell Lineage , Pattern Recognition, Automated , Animals , Apoptosis , Cell Survival , Cells, Cultured , Erythropoietin , Female , Humans , Mice , Mice, Transgenic , Neural Networks, Computer , Phenotype , ROC Curve , Recombinant Proteins , Time Factors
...