Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Nat Biotechnol ; 29(7): 615-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21747388

ABSTRACT

Preclinical and clinical studies conducted in the mid-1990s reported strong association and causality between the T-cell helper (T(H)) 1 inductor cytokine interleukin (IL)-12 and numerous immune-mediated disorders, which spurred the development of therapeutic agents targeting IL-12 function. One of the first to enter the clinic, ustekinumab, is a human monoclonal antibody (mAb) that binds to the p40 subunit of IL-12. Subsequent to the generation of ustekinumab, it was discovered that IL-23 also contains the p40 subunit. Thus, although ustekinumab was designed to target IL-12, it also modulates IL-23, a cytokine important to the development and/or maintenance of T(H)17 cells. Clinical observations established that IL-12/23p40 is integral to the pathologies of psoriasis, psoriatic arthritis and Crohn's disease. The molecular and cellular evaluations conducted in ustekinumab clinical programs have provided numerous insights into the pathologic processes of these disorders, illustrating how a novel molecular entity can contribute to our understanding of disease. The individual contributions of these cytokines to specific pathologies require investigation and clinical evaluation of the role of IL-12- and IL-23-specific inhibitors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Drug Delivery Systems/methods , Interleukin-12/antagonists & inhibitors , Interleukin-23/antagonists & inhibitors , Signal Transduction/physiology , Animals , Antibodies, Monoclonal, Humanized , Humans , Signal Transduction/drug effects , Ustekinumab
3.
J Drugs Dermatol ; 9(6): 677-83, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20645530

ABSTRACT

BACKGROUND: Ustekinumab, a fully human immunoglobulin (Ig) G1K monoclonal antibody directed against the p40 subunit of interleukin (IL)-12/23, has demonstrated efficacy in patients with moderate-to-severe psoriasis. OBJECTIVE: To evaluate the effect of IL-12/23 inhibition on immunocompetency by antigen-recall response in a preclinical multiple-dose toxicology study and three single-dose, phase 1 studies. METHODS: Cynomolgus monkeys (Mauritius; n = 32) treated with subcutaneous (s.c.) placebo or ustekinumab 22.5 or 45 mg/kg twice weekly for 26 weeks were assessed for antibody responsiveness to keyhole limpet hemocyanin (KLH). Patients with psoriasis or multiple sclerosis who received a single-dose of placebo (n = 8) or ustekinumab (n = 46) 0.09-4.5 mg/kg intravenous (i.v.) or 0.27-2.7 mg/kg s.c. were assessed by pneumococcal and tetanus antigen challenge. Primary T-cell response was not assessed in humans. RESULTS: Anti-KLH antibody responses in ustekinumab-treated cynomolgus monkeys were comparable to those observed in placebo-treated animals. A normal antibody response (> or = two-fold increase from baseline) to pneumococcal antigen was seen in 34/46 (73.9%) ustekinumab-treated versus 4/8 (50%) placebo-treated patients. A normal antigen-recall response (> or = four-fold increase from baseline) was seen in 12/20 (60%) ustekinumab- and 4/5 (80%) placebo-treated patients following tetanus toxoid exposure. Percentages of circulating immune cells were not affected by ustekinumab treatment. CONCLUSION: Results in nonhuman primates and human patients suggest that ustekinumab treatment does not significantly impair recall humoral immune system functions.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunity, Humoral/drug effects , Immunologic Memory/drug effects , Animals , Antibodies, Monoclonal, Humanized , Hemocyanins/immunology , Humans , Macaca fascicularis , Pneumococcal Vaccines/immunology , Tetanus Toxoid/immunology , Ustekinumab
4.
Birth Defects Res B Dev Reprod Toxicol ; 89(5): 351-63, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20540088

ABSTRACT

BACKGROUND: Ustekinumab is a human monoclonal antibody that binds to the p40 subunit of interleukin (IL) 12 and IL-23 and inhibits their pharmacological activity. To evaluate potential effects of ustekinumab treatment during pregnancy, developmental studies were conducted in cynomolgus macaques. METHODS: Ustekinumab was tested in two embryo/fetal development (EFD) studies and in a combined EFD/pre and postnatal development (PPND) study. In the EFD studies, pregnant macaques (12/group) were dosed with saline or ustekinumab (9 mg/kg IV, 22.5 mg/kg SC, or 45 mg/kg IV or SC during the period of major organogenesis, gestation day [GD] 20-50). Fetuses were harvested on GD100-102 and examined for any effects on development. In the EFD/PPND study, pregnant macaques were injected with saline or ustekinumab (22.5 or 45 mg/kg SC) from GD20 through lactation day 33. Infants were examined from birth through 6 months of age for morphological and functional development. Potential effects on the immune system were evaluated by immunophenotyping of peripheral blood lymphocytes and immunohistopathology of lymphoid tissues in fetuses and infants and by T-dependent antibody response (TDAR) to KLH and TTX and by DTH response in infants. Ustekinumab concentrations were measured in serum from dams, fetus, and infants and in breast milk. RESULTS: Ustekinumab treatment produced no maternal toxicity and no toxicity in the fetuses or infants, including no effects on the TDAR or DTH responses. Ustekinumab was present in serum from GD100 fetuses and was present in infant serum through day 120 post-birth. Low levels of ustekinumab were present in breast milk. CONCLUSIONS: Exposure of macaque fetuses and infants to ustekinumab had no adverse effects on pre- and postnatal development.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Embryonic Development/drug effects , Lactation/drug effects , Macaca fascicularis/embryology , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibody Formation , Female , Fetus/drug effects , Humans , Immune System/drug effects , Infant , Macaca fascicularis/immunology , Pregnancy , Ustekinumab
5.
Birth Defects Res B Dev Reprod Toxicol ; 89(2): 116-23, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20151458

ABSTRACT

BACKGROUND: Intetumumab is a human IgG1 anti-alphav-integrin monoclonal antibody that inhibits angiogenesis. Integrin binding and angiogenesis are important in reproduction including fertilization, implantation, and embryofetal development. These studies were designed to determine the pharmacological relevance of the rabbit for the evaluation of potential effects on embryofetal development and to evaluate the placental transfer of intetumumab in rabbits. METHODS: In vitro pharmacology studies evaluated the binding of intetumumab to rabbit cells and the inhibition of vessel sprouting from rabbit aorta. For the evaluation of placental transfer, pregnant rabbits (8/group) were injected intravenously with intetumumab 50 or 100 mg/kg every 2 days from Gestation Day (GD)7 to GD19. Maternal sera, fetal homogenates/sera, and amniotic fluid were collected at necropsy on GD19 or GD28 for evaluation of intetumumab concentrations. Clinical condition of the dams was monitored and fetuses were screened for abnormalities. RESULTS: Intetumumab (5-40 microg/mL) inhibited aortic cell adhesion to vitronectin and vessel sprouting from rabbit aortic rings. Immunohistochemical staining of rabbit tissues demonstrated binding of intetumumab to placenta. Administration of intetumumab to pregnant rabbits was well tolerated by the dams and the fetuses did not show major abnormalities. Fetal exposure to intetumumab relative to maternal exposure was <0.1% on GD19 and 100-130% on GD29. CONCLUSIONS: The rabbit is a pharmacologically relevant species for evaluation of potential developmental effects of intetumumab. Intetumumab crosses the rabbit placenta during the fetal period (GD 19-28).


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Integrin alpha5/immunology , Maternal-Fetal Exchange/drug effects , Placenta/drug effects , Amniotic Fluid/drug effects , Amniotic Fluid/metabolism , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Blood Vessels/drug effects , Blood Vessels/growth & development , Cell Adhesion/drug effects , Cells, Cultured , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Embryonic Development/physiology , Endothelial Cells/drug effects , Female , Fetal Development/drug effects , Fetal Development/physiology , Fetus/drug effects , Fetus/metabolism , Injections, Intravenous , Maternal Exposure , Maternal-Fetal Exchange/physiology , Neovascularization, Physiologic/drug effects , Placenta/metabolism , Pregnancy , Rabbits
6.
Expert Rev Clin Immunol ; 5(5): 499-521, 2009 Sep.
Article in English | MEDLINE | ID: mdl-20477639

ABSTRACT

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.

7.
Int J Toxicol ; 27(4): 341-7, 2008.
Article in English | MEDLINE | ID: mdl-18821398

ABSTRACT

Monoclonal antibodies directed against tumor necrosis factor alpha (TNFalpha) are currently employed in the treatment of various immune-mediated diseases. These studies were designed to evaluate potential effects of anti-TNFalpha treatment in mice during pregnancy and lactation on the development of the immune system in the F1 generation. Pregnant CD-1 mice were treated with vehicle or with 10 or 40 mg/kg of an anti-mouse TNFalpha monoclonal antibody (mAb) (cV1q) on days 6, 12, and 18 of gestation and on days 3, 9, and 15 of lactation. Evaluation of immune system functionality was conducted in F1 generation mice at 11 weeks of age. Immune function was evaluated by splenocyte phenotyping, immunoglobulin M (IgM) antibody response to sheep red blood cells (SRBCs), spleen cell proliferative response to anti-CD3, and natural killer cell activity. Treatment of pregnant mice with cV1q produced no adverse effects in the dams and no adverse effects in the F1 generation. In general, the functioning of the immune system of the F1 generation did not appear to be adversely affected following exposure to cV1q in utero and during lactation. The only statistically significant change was a slight (approximately 20%) reduction in the spleen cell expansion in response to SRBC immunization in the female F1 mice from the 40 mg/kg cV1q treatment group. In conclusion, administration of a monoclonal antibody against mouse TNFalpha during pregnancy and lactation had little or no effect on selected immune parameters in mice, with only a possible minor attenuation of spleen cell response to immunization noted in the female F1 generation at 11 weeks of age.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Immune System/embryology , Immune System/growth & development , Lactation , Tumor Necrosis Factor-alpha/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Formation , Female , Immunoglobulin M/immunology , Immunophenotyping , Killer Cells, Natural/immunology , Male , Mice , Pregnancy
8.
Am J Reprod Immunol ; 58(2): 138-49, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17631007

ABSTRACT

PROBLEM: The use of anti-TNF-alpha therapies during pregnancy and lactation on the development of the neonatal immune system has not been fully established. The purpose of this study was to evaluate whether treatment of macaques with an anti-TNF-alpha monoclonal antibody (golimumab) during pregnancy and lactation would result in defects in the developing immune system. METHOD OF STUDY: Pregnant macaques were treated with golimumab during pregnancy and lactation. Immune system development was evaluated by histopathology, lymphocyte subset analysis and functional challenging of the infant immune system (humoral immune response to KLH and TTX, and DTH skin reaction). RESULTS: In utero and postnatal exposure to golimumab had no effect on T and B cell populations in blood and lymphoid tissues and did not impair the ability of the infants to mount an immune response to antigen challenge. CONCLUSION: Treatment of pregnant macaques with golimumab throughout pregnancy and lactation did not affect the development and maturation of the immune system in the offspring.


Subject(s)
Antibodies, Monoclonal/physiology , Immune System/immunology , Lactation/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Embryonic Development/immunology , Female , Fetal Development/immunology , Macaca fascicularis , Male , Pregnancy
9.
Clin Cancer Res ; 11(19 Pt 1): 6959-65, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16203788

ABSTRACT

PURPOSE: CNTO 95 is a fully human anti-alphav integrin monoclonal antibody that inhibits macaque and rodent angiogenesis and inhibits human tumor growth in rodents. The purpose of these studies was to evaluate the preclinical safety of long-term administration of CNTO 95 in cynomolgus macaques. EXPERIMENTAL DESIGN: The in vitro binding profiles of CNTO 95 to human and macaque tissues and the in vivo binding to macaque tissues was evaluated by immunohistochemistry. The preclinical safety of CNTO 95 (10 and 50 mg/kg, i.v.) was evaluated in macaques treated once per week for up to 6 months. Safety was evaluated by clinical observations, ophthalmic and physical examinations (including heart rate, blood pressure, and electrocardiogram), clinical pathology (including coagulation parameters), and comprehensive anatomic pathology. The effect of CNTO 95 (50 mg/kg, i.v.) on incisional wound healing was evaluated in macaques. RESULTS: The tissue binding studies showed that CNTO 95 bound with mild to moderate intensity to macaque and human endothelial cells, epithelial cells, and vascular smooth muscle cells in most normal tissues examined. CNTO 95 showed strong to intense staining to the positive control tissue, human placenta. Despite the widespread binding to normal tissues, treatment of cynomolgus macaques with CNTO 95 produced no signs of toxicity and no histopathologic changes in any of the tissues examined (including ovaries and bone growth plates). CNTO 95 did not impair wound healing. CONCLUSION: These studies show that CNTO 95 is safe and, unlike some other angiogenesis inhibitors, does not seem to inhibit normal physiologic angiogenesis.


Subject(s)
Antibodies, Monoclonal/chemistry , Integrin alphaV/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Aorta/metabolism , Area Under Curve , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Female , Humans , Immune System , Immunohistochemistry , Macaca fascicularis , Male , Myocytes, Smooth Muscle/cytology , Neovascularization, Pathologic , Placenta/metabolism , Protein Binding , Time Factors , Wound Healing
10.
J Immunol ; 175(7): 4761-8, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16177124

ABSTRACT

IL-12p40 is a shared subunit of two cytokines with overlapping activities in the induction of autoreactive Th1 cells and therefore a potential target of therapy in Th1-mediated diseases. We have examined whether ongoing disease in a nonhuman primate model of multiple sclerosis (MS) can be suppressed with a new human IgG1kappa Ab against human IL-12p40. Lesions developing in the brain white matter were visualized and characterized with standard magnetic resonance imaging techniques. To reflect the treatment of MS patients, treatment with the Ab was initiated after active brain white matter lesions were detected in T2-weighted images. In placebo-treated control monkeys we observed the expected progressive increase in the total T2 lesion volume and markedly increased T2 relaxation times, a magnetic resonance imaging marker of inflammation. In contrast, in monkeys treated with anti-IL-12p40 Ab, changes in the total T2 lesion volume and T2 relaxation times were significantly suppressed. Moreover, the time interval to serious neurological deficit was delayed from 31 +/- 10 to 64 +/- 20 days (odds ratio, 0.312). These results, in a disease model with high similarity to MS, are important for ongoing and planned trials of therapies that target IL-12 and/or IL-23.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Callithrix , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interleukin-12/immunology , Protein Subunits/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Brain/pathology , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Interleukin-12 Subunit p40 , Magnetic Resonance Imaging , Male , Multiple Sclerosis/diagnosis , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Multiple Sclerosis/prevention & control
11.
J Immunotoxicol ; 1(3): 131-9, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-18958646

ABSTRACT

Inhibitors of tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) have been shown to be efficacious in a number of autoimmune diseases. In this study, the safety of long-term administration of anti-TNF-alpha and anti-IL-6 monoclonal antibodies (mAbs) was evaluated in cynomolgus macaques. Effects on the immune system were evaluated by analysis of lymphocyte subsets and histopathology of lymphoid tissues. To evaluate the functioning of the immune system, the ability of mAb-treated monkeys to mount a humoral immune response (IgG and IgM) to keyhole limpet hemocyanin (KLH) was evaluated. Treatment with the anti-TNF-alpha mAb produced no histopathological changes in any of the lymphoid tissues examined. There was a small (< 2-fold) elevation in circulating T-and B-lymphocytes during anti-TNF-alpha mAb treatment that was not considered to be toxicologically significant. The antibody response to KLH was unaffected by anti-TNF-alpha mAb treatment. Treatment with anti-IL-6 mAb resulted in a decrease in the size of germinal centers in the spleens of a minority of the animals and a modest but significant decrease in the IgG antibody response to KLH. Weekly intravenous treatment with the anti-IL-6 mAb and twice-weekly subcutaneous treatment with the anti-TNF-alpha mAb for up to 6 months was not associated with any signs of toxicity, and no animal developed an infection throughout the study period. This study demonstrates that the anti-IL-6 and anti-TNF-alpha mAbs produce specific modulating effects on the immune system without rendering the animals immune compromised.

12.
Curr Opin Mol Ther ; 6(1): 10-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15011776

ABSTRACT

Despite advances at the level of gene sequence (eg, humanized versus murine antibodies), expression systems (eg, mammalian versus prokaryotic) and post-expression modification (eg, 'PEGylation'), clinical immunogenicity of therapeutic proteins remains a concern. Although animals are routinely and effectively used to evaluate pharmacological activity and to support a claim of safety for recombinant proteins, their usefulness for predicting clinical immunogenicity is more questionable. This review argues that recombinant proteins can be grouped into immunogenic classes; for some of these classes, for example, bacterial proteins, immunogenicity in animals is often predictive for humans, but for others, for example, fully human proteins, even data from non-human primates can have little predictive value. We will attempt to make the case that for a variety of immunological and practical constraints, animal studies, even those conducted in non-human primates, have limited predictive power for immunogenicity in humans, and tend to over-predict clinical immunogenicity.


Subject(s)
Antigens/therapeutic use , Drug Evaluation, Preclinical/methods , Immune System/drug effects , Predictive Value of Tests , Recombinant Proteins/immunology , Animals , Animals, Laboratory , Antigens/administration & dosage , Antigens/immunology , Humans , Recombinant Proteins/administration & dosage
13.
Platelets ; 15(7): 409-18, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15745312

ABSTRACT

Monoclonal antibodies have been firmly established as human therapeutics. Their high affinity and specificity for target antigens minimize adverse reactions and their molecular size results in extended circulation times relative to small molecule pharmaceuticals. The ability to customize the pharmacokinetics in a rational manner can enhance the potential for these and other classes of biologicals. We have systematically studied the effect of site-specific pegylation of the Fab' fragment of the anti-GPIIb/IIIa, alphavbeta3 antibody c7E3. Regardless of the molecular weight of the PEG molecules, the intrinsic affinity of the resulting constructs remained unchanged. However, in functional assays measuring inhibition of platelet aggregation, the calculated IC50 values of the conjugates decreased with increasing molecular weight of the conjugated PEG. It was determined that the molecular size of the conjugates affects antigen accessibility and whereas high levels of binding to antigen molecules on cells with high antigen density can be demonstrated with the Fab fragment, comparable levels are not achievable with large molecular weight conjugates. In spite of the inability of the larger PEG constructs to achieve saturation binding, functional inhibition of platelet aggregation consistent with saturation binding was demonstrated and the increased molecular size of the conjugates led to predictably prolonged inhibition of platelet aggregation.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoglobulin Fab Fragments/chemistry , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation/drug effects , Polyethylene Glycols/chemistry , Abciximab , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Line , Haplorhini , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/pharmacology , Mice , Mice, Inbred BALB C , Platelet Aggregation Inhibitors/immunology , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors
14.
J Immunol ; 169(11): 6554-63, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12444167

ABSTRACT

The experimental autoimmune encephalomyelitis (EAE) model in the common marmoset approximates recognized features of the human disease multiple sclerosis (MS) with regard to its clinical presentation as well as neuropathological and radiological aspects of the lesions in brain and spinal cord. IL-12 is a proinflammatory cytokine that is produced by APC and promotes differentiation of Th1 effector cells. IL-12 is produced in the developing lesions of patients with MS as well as in EAE-affected animals. Previously it was shown that interference in IL-12 pathways effectively prevents EAE in rodents. In this study we report that in vivo neutralization of IL-12p40 using a novel Ab has beneficial effects in the myelin-induced EAE model in common marmosets. The Ab was injected i.v. at 7-day intervals starting well after immunization (day 14) and was continued until the end of the study (day 86). Stable levels of the Ab were measured 3 days after each injection throughout the study period. During this period anti-Ab responses could not be detected. We demonstrate that anti-IL-12p40 treatment has a protective effect on the neurological dysfunction as well as on neuropathological changes normally observed in the brain and spinal cord of EAE-affected individuals.


Subject(s)
Antibodies, Monoclonal/pharmacology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interleukin-12/antagonists & inhibitors , Protein Subunits/antagonists & inhibitors , Animals , Antibodies, Anti-Idiotypic/biosynthesis , Antibodies, Monoclonal/administration & dosage , B-Lymphocytes/immunology , Brain/immunology , Brain/pathology , Callithrix , Cross Reactions , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Interleukin-12 Subunit p40 , Multiple Sclerosis/etiology , Multiple Sclerosis/immunology , Myelin Proteins/immunology , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/immunology
15.
Arthritis Res ; 4(5): R7, 2002.
Article in English | MEDLINE | ID: mdl-12223110

ABSTRACT

Anti-tumor-necrosis-factor-alpha (TNF-alpha) monoclonal antibody was used to treat Tg197 transgenic mice, which constitutively produce human TNF-alpha (hTNF-alpha) and develop a progressive polyarthritic disease. Treatment of both young (7- or 8-week-old) and aged (27- or 28-week-old) mice commenced when at least two limbs showed signs of moderate to severe arthritis. The therapeutic efficacy of anti-TNF-alpha antibody was assessed using various pathological indicators of disease progression. The clinical severity of arthritis in Tg197 mice was significantly reduced after anti-TNF-alpha treatment in comparison with saline-treated mice and in comparison with baseline assessments in both young and aged mice. The treatment with anti-TNF-alpha prevented loss of body weight. Inflammatory pathways as reflected by elevated circulating hTNF-alpha and local expression of various proinflammatory mediators were all diminished by anti-TNF-alpha treatment, confirming a critical role of hTNF-alpha in this model of progressive polyarthritis. More importantly, the amelioration of the disease was associated with reversal of existing structural damage, including synovitis and periosteal bone erosions evident on histology. Repair of cartilage was age dependent: reversal of cartilage degradation after anti-TNF-alpha treatment was observed in young mice but not in aged mice.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/therapy , Joints/pathology , Tumor Necrosis Factor-alpha/immunology , Animals , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Chemokines/genetics , Chemokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Forearm/pathology , Heterozygote , Hindlimb/pathology , Joints/drug effects , Mice , Mice, Transgenic , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...