1
|
Zhu Y, Zhuang W, Cheng H. Strategies to Enhance Protein Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:6457-6470. [PMID: 40052814 PMCID: PMC11924232 DOI: 10.1021/acs.langmuir.4c04636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/19/2025]
Abstract
Therapeutic proteins play a crucial role in modern healthcare. However, the rapid clearance of proteins in the circulation system poses a significant threat to their therapeutic efficacy. The generation of anti-drug antibodies expedites drug clearance, resulting in another challenge to overcome in protein delivery. Several methods to increase the circulation half-lives of these proteins and to minimize their immunogenicity have been developed. This Review discusses the causes of protein clearance in the body, evaluates the FDA-approved strategies to prolong protein circulation, and highlights recent progress in the field. Additionally, the strengths and drawbacks of these methods and our perspectives for advancing protein delivery are provided.
Collapse
Affiliation(s)
- Yucheng Zhu
- Department of Materials Science
and Engineering, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Weisi Zhuang
- Department of Materials Science
and Engineering, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Hao Cheng
- Department of Materials Science
and Engineering, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
2
|
Sandeep, Shinde SH, Pande AH. Polyspecificity - An emerging trend in the development of clinical antibodies. Mol Immunol 2023; 155:175-183. [PMID: 36827806 DOI: 10.1016/j.molimm.2023.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/01/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023]
Abstract
The essence of the growth and development of therapeutic conventional monoclonal antibodies (MAbs) for the treatment of various disorders is the aptitude of MAbs to precisely bind a target antigen and neutralise or promote its activity. However, the conventional antibodies are monoclonal i.e., both paratopes bind to the same epitope. But most of the pathophysiological conditions are multifaceted, hence targeting/blocking/inhibition of more than one epitope/antigen is more promising than one epitope/antigen. Polyspecific antibodies (PsAbs) have the potential to concurrently bind to more than one target and are the next-generation antibodies that augment efficacy in both clinical and non-clinical contexts. Thus, the trend of engineering and developing various formats of PsAbs is emerging. In this review, we have briefly discussed the importance of antibody polyspecificity and PsAbs approved for clinical use. Subsequently, we have discussed the role of TNF-α and IL-23 in inflammatory diseases and stressed the need for developing anti-TNF-α and anti-IL-23 bispecific antibodies.
Collapse
Affiliation(s)
- Sandeep
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Suraj H Shinde
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
3
|
Zaman R, Islam RA, Ibnat N, Othman I, Zaini A, Lee CY, Chowdhury EH. Current strategies in extending half-lives of therapeutic proteins. J Control Release 2019; 301:176-189. [PMID: 30849445 DOI: 10.1016/j.jconrel.2019.02.016] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 01/23/2023]
Abstract
Macromolecular protein and peptide therapeutics have been proven to be effective in treating critical human diseases precisely. Thanks to biotechnological advancement, a huge number of proteins and peptide therapeutics were made their way to pharmaceutical market in past few decades. However, one of the biggest challenges to be addressed for protein therapeutics during clinical application is their fast degradation in serum and quick elimination owing to enzymatic degradation, renal clearance, liver metabolism and immunogenicity, attributing to the short half-lives. Size and hydrophobicity of protein molecules make them prone to kidney filtration and liver metabolism. On the other hand, proteasomes responsible for protein destruction possess the capability of specifically recognizing almost all kinds of foreign proteins while avoiding any unwanted destruction of cellular components. At present almost all protein-based drug formulations available in market are administered intravenously (IV) or subcutaneously (SC) with high dosing at frequent interval, eventually creating dose-fluctuation-related complications and reducing patient compliance vastly. Therefore, artificially increasing the therapeutic half-life of a protein by attaching to it a molecule that increases the overall size (eg, PEG) or helps with receptor mediated recycling (eg, albumin), or manipulating amino acid chain in a way that makes it more prone towards aggregate formation, are some of the revolutionary approaches to avoid the fast degradation in vivo. Half-life extension technologies that are capable of dramatically enhancing half-lives of proteins in circulation (2-100 folds) and thus improving their overall pharmacokinetic (PK) parameters have been successfully applied on a wide range of protein therapeutics from hormones and enzymes, growth factor, clotting factor to interferon. The focus of the review is to assess the technological advancements made so far in enhancing circulatory half-lives and improving therapeutic potency of proteins.
Collapse
Affiliation(s)
- Rahela Zaman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Rowshan Ara Islam
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Nabilah Ibnat
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Anuar Zaini
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Chooi Yeng Lee
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
4
|
A New Venue of TNF Targeting. Int J Mol Sci 2018; 19:ijms19051442. [PMID: 29751683 PMCID: PMC5983675 DOI: 10.3390/ijms19051442] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/25/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
The first Food and Drug Administration-(FDA)-approved drugs were small, chemically-manufactured and highly active molecules with possible off-target effects, followed by protein-based medicines such as antibodies. Conventional antibodies bind a specific protein and are becoming increasingly important in the therapeutic landscape. A very prominent class of biologicals are the anti-tumor necrosis factor (TNF) drugs that are applied in several inflammatory diseases that are characterized by dysregulated TNF levels. Marketing of TNF inhibitors revolutionized the treatment of diseases such as Crohn’s disease. However, these inhibitors also have undesired effects, some of them directly associated with the inherent nature of this drug class, whereas others are linked with their mechanism of action, being pan-TNF inhibition. The effects of TNF can diverge at the level of TNF format or receptor, and we discuss the consequences of this in sepsis, autoimmunity and neurodegeneration. Recently, researchers tried to design drugs with reduced side effects. These include molecules with more specificity targeting one specific TNF format or receptor, or that neutralize TNF in specific cells. Alternatively, TNF-directed biologicals without the typical antibody structure are manufactured. Here, we review the complications related to the use of conventional TNF inhibitors, together with the anti-TNF alternatives and the benefits of selective approaches in different diseases.
Collapse
|
5
|
Forest T, Xu Q, Kuruvilla S, Vu H, Vlasakova K, Glaab WE, Hines C, Xun S. Magnetic Resonance and Ultrastructural Characterization of PEGylation-associated Vacuolation in Nonclinical Models. Toxicol Pathol 2017; 45:604-613. [PMID: 28673196 DOI: 10.1177/0192623317714068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Conjugation with polyethylene glycol (PEG) is a strategy for improving the pharmaceutical properties of therapeutic proteins. In nonclinical studies of PEGylated compounds, microscopic tissue vacuolation is often observed, characterized ultrastructurally in this report by lysosomal distension. Although PEGylation-associated vacuolation appears to be of limited toxicologic concern when alternative therapies are limited, the risk-benefit considerations may be impacted by uncertainty about reversibility, lack of methods for monitoring PEG accumulation in vivo without biopsy, and the variability in tissues affected depending on species studied. We demonstrate the use of magnetic resonance spectroscopy (MRS) to measure PEG concentrations at multiple time points in vivo in the kidney with comparison to PEG concentrations ex vivo in body fluids and tissue extracts using nuclear magnetic resonance (NMR) spectroscopy. Furthermore, we demonstrate the use of these techniques to study distribution and elimination of PEG in a dog model of PEGylation-associated vacuolation. This report suggests that MRS could be further investigated as a feasible imaging-based method for monitoring PEG accumulation in a clinical setting in conjunction with NMR quantitation of PEG in plasma and urine.
Collapse
Affiliation(s)
- Thomas Forest
- Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Qiuwei Xu
- Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Sabu Kuruvilla
- Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Heather Vu
- Merck Research Laboratories, West Point, Pennsylvania, USA
| | | | | | - Cathy Hines
- Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Song Xun
- Merck Research Laboratories, West Point, Pennsylvania, USA
| |
Collapse
|
6
|
Sedger LM, McDermott MF. TNF and TNF-receptors: From mediators of cell death and inflammation to therapeutic giants - past, present and future. Cytokine Growth Factor Rev 2014; 25:453-72. [PMID: 25169849 DOI: 10.1016/j.cytogfr.2014.07.016] [Citation(s) in RCA: 566] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tumor Necrosis Factor (TNF), initially known for its tumor cytotoxicity, is a potent mediator of inflammation, as well as many normal physiological functions in homeostasis and health, and anti-microbial immunity. It also appears to have a central role in neurobiology, although this area of TNF biology is only recently emerging. Here, we review the basic biology of TNF and its normal effector functions, and discuss the advantages and disadvantages of therapeutic neutralization of TNF - now a commonplace practice in the treatment of a wide range of human inflammatory diseases. With over ten years of experience, and an emerging range of anti-TNF biologics now available, we also review their modes of action, which appear to be far more complex than had originally been anticipated. Finally, we highlight the current challenges for therapeutic intervention of TNF: (i) to discover and produce orally delivered small molecule TNF-inhibitors, (ii) to specifically target selected TNF producing cells or individual (diseased) tissue targets, and (iii) to pre-identify anti-TNF treatment responders. Although the future looks bright, the therapeutic modulation of TNF now moves into the era of personalized medicine with society's challenging expectations of durable treatment success and of achieving long-term disease remission.
Collapse
Affiliation(s)
- Lisa M Sedger
- Australian School of Advanced Medicine, Macquarie University, North Ryde, NSW 2109, Australia; The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia.
| | - Michael F McDermott
- Experimental Rheumatology, National Institute for Health Research - Leeds Musculoskeletal Biomedical Research Unit (NIHR-LMBRU), and Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, St James University, Beckett Street, West Yorkshire, Leeds LS9 7TF, UK.
| |
Collapse
|
7
|
He ZY, Chu BY, Wei XW, Li J, Edwards CK, Song XR, He G, Xie YM, Wei YQ, Qian ZY. Recent development of poly(ethylene glycol)-cholesterol conjugates as drug delivery systems. Int J Pharm 2014; 469:168-78. [DOI: 10.1016/j.ijpharm.2014.04.056] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 04/15/2014] [Accepted: 04/23/2014] [Indexed: 11/28/2022]
|
8
|
McDonnell T, Ioannou Y, Rahman A. PEGylated drugs in rheumatology--why develop them and do they work? Rheumatology (Oxford) 2013; 53:391-6. [PMID: 23962623 PMCID: PMC3930883 DOI: 10.1093/rheumatology/ket278] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Lack of efficacy and drug-related adverse effects are important reasons for the discontinuation of treatment in patients with rheumatic diseases. The development of new biologic therapies seeks to address these problems by specifically targeting the pathogenic mechanisms of disease. Most current biologics are proteins (particularly antibodies and enzymes) administered parenterally. It is important to optimize properties such as serum half-life, immunogenicity and solubility. Companies have thus begun to modify the drugs by conjugate chemistry, binding inert molecules such as polyethylene glycol (PEG) to biologic molecules to improve their pharmacodynamic properties. The use of PEG to alter these properties has to be weighed against the negative aspects of PEGylation, such as decreased activity and heterogeneity. This review focuses on the currently available PEGylated drugs used in rheumatological diseases, their efficacy, drawbacks and the current clinical trial evidence supporting their use.
Collapse
Affiliation(s)
- Thomas McDonnell
- Centre for Rheumatology Research, Division of Medicine, University College London, Room 412 Rayne Institute, 5 University Street, London WC1E 6JF, UK.
| | | | | |
Collapse
|
9
|
Ware CF. Protein therapeutics targeted at the TNF superfamily. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2013; 66:51-80. [PMID: 23433455 DOI: 10.1016/b978-0-12-404717-4.00002-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Protein-based drugs with their unequivocal specificity achieved the long sought milestone of selectively disrupting cytokine pathways to alleviate ongoing inflammation. Tumor necrosis factor (TNF), a member of the superfamily of cytokines involved in regulating immune and inflammatory processes, provides an exemplary model of protein therapeutics. Antibody and receptor-based inhibitors of TNF modify inflammation leading to dramatic improvement in patients with certain autoimmune diseases. Collectively, the structure, specificity and valence of these protein-based drugs provide direct evidence that the essential mechanism of action is antagonism of the ligand-receptor interaction. Accumulating clinical knowledge regarding TNF inhibitors also provide insights into the mechanisms involved in different autoimmune diseases. Experience in the development of an arsenal of biologics directed at TNF has additionally contributed to knowledge toward overcoming the challenges of protein drugs, which include production, delivery, antigenicity and pharmacodynamics. Dramatic clinical outcomes with TNF inhibitors are driving investigation and development of biologics toward other members of the TNF superfamily to selectively alter functional properties of the immune system.
Collapse
Affiliation(s)
- Carl F Ware
- Laboratory of Molecular Immunology, Infectious and Inflammatory Diseases Center, Sanford Burnham Medical Research Institute, La Jolla, CA, USA.
| |
Collapse
|
10
|
|
11
|
Therrien FJ, Agharazii M, Lebel M, Larivière R. Neutralization of tumor necrosis factor-alpha reduces renal fibrosis and hypertension in rats with renal failure. Am J Nephrol 2012; 36:151-61. [PMID: 22813949 DOI: 10.1159/000340033] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 06/12/2012] [Indexed: 12/18/2022]
Abstract
BACKGROUND Increased production of tumor necrosis factor-α (TNF-α) in chronic kidney disease may be involved in the progression of renal failure and injury, and cardiovascular disease. We investigated the effect of TNF-α neutralization on renal failure, inflammation and fibrosis, and blood pressure in rats with renal failure. METHODS AND RESULTS Renal failure was induced by renal mass reduction and the animals were treated with PEG-sTNFR1, a pegylated form of soluble TNF type 1 receptor that neutralizes TNF-α, for 6 weeks. Systolic, diastolic and mean arterial pressures were higher in renal failure rats that were associated with increased serum creatinine, albuminuria and renal injury comprised of blood vessel media hypertrophy, focal and segmental glomerulosclerosis, tubular atrophy and interstitial inflammation and fibrosis. These changes were associated with greater levels of TNF-α, transforming growth factor (TGF)-β1, nuclear transcription factor NF-ĸB and cytosolic phospho-IĸB-α, and inflammatory markers expression (ICAM-1, VCAM-1 and MCP-1). Moreover, endothelin (ET)-1 production was also increased, whereas nitric oxide (NO) release was decreased. TNF-α neutralization reduced hypertension, albuminuria and renal inflammation and fibrosis, which were coupled to a reduction in renal NF-ĸB activation, inflammatory markers expression, TGF-β1 and ET-1 production, and an increase in NO release. CONCLUSION Neutralization of TNF-α in rats with renal failure decreases NF-ĸB activity that is associated with a reduction in renal TGF-β1 and ET-1 production, and an improvement of NO release. These effects likely reduce renal inflammation and fibrosis, and blood pressure indicating a pivotal role for TNF-α, at least, in the progression of renal injury.
Collapse
Affiliation(s)
- Frédérick J Therrien
- Nephrology and Hypertension, CHUQ Research Centre, L'Hôtel-Dieu de Québec Hospital and Department of Medicine, Université Laval, Quebec, Que., Canada
| | | | | | | |
Collapse
|
12
|
Zhang C, Fan K, Luo H, Ma X, Liu R, Yang L, Hu C, Chen Z, Min Z, Wei D. Characterization, efficacy, pharmacokinetics, and biodistribution of 5kDa mPEG modified tetrameric canine uricase variant. Int J Pharm 2012; 430:307-17. [DOI: 10.1016/j.ijpharm.2012.03.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/17/2012] [Accepted: 03/31/2012] [Indexed: 02/07/2023]
|
13
|
Byrnes-Blake KA, Pederson S, Klucher KM, Anderson-Haley M, Miller DM, Lopez-Talavera JC, Freeman JA. Pharmacokinetics and pharmacodynamics of pegylated interferon lambda-1 in cynomolgus monkeys. J Interferon Cytokine Res 2012; 32:198-206. [PMID: 22280056 DOI: 10.1089/jir.2011.0075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Type III lambda interferons (IFNs) have activity similar to type I IFNs, but a more restricted receptor distribution. A pegylated human IFN lambda-1 (pegIFNλ) is under development for chronic hepatitis C. Induction of receptor signaling (STAT1 phosphorylation) and expression of interferon-stimulated genes (ISGs) by pegIFNλ were assessed in, respectively, cynomolgus monkey leukocyte subsets and hepatocytes stimulated in vitro. ISG induction by pegIFNλ or IFNα was also assessed in peripheral leukocytes and liver biopsies after single and repeat (x3) dosing of pegIFNλ (0.03, 0.3, 3.0 mg/kg) or unpegylated IFNα-2b (10(7) IU/kg). Single-dose pharmacokinetics of pegIFNλ were evaluated. Strong ISG induction occurred in cultured hepatocytes and liver biopsies with both pegIFNλ and IFNα. However, STAT1 phosphorylation, MHC class 1 upregulation, and ISG induction in leukocytes only occurred with IFNα. Serum neopterin was unaffected by pegIFNλ; however, β-2-microglobulin was elevated at all doses. The terminal half-life of pegIFNλ was 23 h with a 59 mL/kg volume of distribution, consistent with other pegylated IFNs. Serum exposure was dose-proportional across the dosing range. These data demonstrate the suitability of cynomolgus monkeys for the preclinical evaluation of pegIFNλ. Additionally, the absence of pegIFNλ pharmacologic activity in leukocytes is consistent with its low receptor expression in blood.
Collapse
Affiliation(s)
- Kelly A Byrnes-Blake
- Department of Preclinical Development, ZymoGenetics, Inc., Seattle, WA 98102, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Kim TH, Choi SJ, Lee YH, Song GG, Ji JD. Soluble triggering receptor expressed on myeloid cells-1 as a new therapeutic molecule in rheumatoid arthritis. Med Hypotheses 2011; 78:270-2. [PMID: 22100630 DOI: 10.1016/j.mehy.2011.10.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 07/15/2011] [Accepted: 10/27/2011] [Indexed: 10/15/2022]
Abstract
Triggering receptor expressed on myeloid cells-1 (TREM-1) is a recently identified cell surface receptor that is expressed mainly on monocytes and neutrophils, and plays an important role as an amplifier of inflammatory response in acute and chronic inflammatory conditions. Recent studies suggested that TREM-1 contributes to the pathogenesis of rheumatoid arthritis (RA) and therefore TREM-1 could be a new therapeutic target in RA. In addition to its membrane-bound form, a soluble form of TREM-1 (sTREM-1) exists that is liberated by the proteolytic cleavage of membrane-bound form. This soluble form works as decoy receptor to prevent the binding of its ligand to membrane-bound TREM-1 and to inhibit the effect of TREM-1 activation. Proteolytic cleavage of TNF receptor (TNFR) has been reported and soluble TNFR are capable of binding and neutralizing TNF, thus working as natural TNF antagonist. Currently, etanercept, a soluble TNF-receptor fusion protein has been widely used to treat RA. In this report, we suggest that sTREM-1 can be used as a new therapeutic molecule in RA.
Collapse
Affiliation(s)
- Tae-Hwan Kim
- The Hospital for Rheumatic Diseases, Hanyang University, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
15
|
Sinclair SM, Shamji MF, Chen J, Jing L, Richardson WJ, Brown CR, Fitch RD, Setton LA. Attenuation of inflammatory events in human intervertebral disc cells with a tumor necrosis factor antagonist. Spine (Phila Pa 1976) 2011; 36:1190-6. [PMID: 21217452 PMCID: PMC3378380 DOI: 10.1097/brs.0b013e3181ebdb43] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN The inflammatory responses of primary human intervertebral disc (IVD) cells to tumor necrosis factor α (TNF-α) and an antagonist were evaluated in vitro. OBJECTIVE To investigate an ability for soluble TNF receptor type II (sTNFRII) to antagonize TNF-α-induced inflammatory events in primary human IVD cells in vitro. SUMMARY OF BACKGROUND DATA TNF-α is a known mediator of inflammation and pain associated with radiculopathy and IVD degeneration. sTNFRs and their analogues are of interest for the clinical treatment of these IVD pathologies, although information on the effects of sTNFR on human IVD cells remains unknown. METHODS IVD cells were isolated from surgical tissues procured from 15 patients and cultured with or without 1.4 nmol/L TNF-α (25 ng/mL). Treatment groups were coincubated with varying doses of sTNFRII (12.5-100 nmol/L). Nitric oxide (NO), prostaglandin E₂ (PGE₂), and interleukin-6 (IL6) levels in media were quantified to characterize the inflammatory phenotype of the IVD cells. RESULTS Across all patients, TNF-α induced large, statistically significant increases in NO, PGE₂, and IL6 secretion from IVD cells compared with controls (60-, 112-, and 4-fold increases, respectively; P < 0.0001). Coincubation of TNF-α with nanomolar doses of sTNFRII significantly attenuated the secretion of NO and PGE₂ in a dose-dependent manner, whereas IL6 levels were unchanged. Mean IC₅₀ values for NO and PGE₂ were found to be 35.1 and 20.5 nmol/L, respectively. CONCLUSION Nanomolar concentrations of sTNFRII were able to significantly attenuate the effects of TNF-α on primary human IVD cells in vitro. These results suggest this sTNFR to be a potent TNF antagonist with potential to attenuate inflammation in IVD pathology.
Collapse
Affiliation(s)
| | | | - Jun Chen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Liufang Jing
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - William J. Richardson
- Department of Surgery, Division of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Christopher R. Brown
- Department of Surgery, Division of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Robert D. Fitch
- Department of Surgery, Division of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Lori A. Setton
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Surgery, Division of Orthopaedic Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
16
|
ZNAMENSKAYA LF, YEGOROVA YUYU, ZITNER SV. Mechanism of the biological effect of the tumor necrosis factor-аlpha at psoriasis. VESTNIK DERMATOLOGII I VENEROLOGII 2011. [DOI: 10.25208/vdv974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Among different cytokines, tumor necrosis factor-аlpha (tumor necrosis factor α, TNF-α) plays a special role in psoriatic immunopathogenesis. Data on this cytokine collected for the recent decades made it possible to create a number of biological drugs blocking TNF-α, which are successfully applied in clinical practice for treating medium to severe psoriasis and psoriatic arthritis. This review presents general information about the cytokine structure and its receptor apparatus, regulation mechanisms of TNF-α synthesis and ways of signal transmission as the basis needed to implement the biological effects of cytokine in the development of psoriatic skin affections.
Collapse
|
17
|
Nakamura K, Radhakrishnan K, Wong YM, Rockson SG. Anti-inflammatory pharmacotherapy with ketoprofen ameliorates experimental lymphatic vascular insufficiency in mice. PLoS One 2009; 4:e8380. [PMID: 20027220 PMCID: PMC2791214 DOI: 10.1371/journal.pone.0008380] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2009] [Accepted: 11/25/2009] [Indexed: 11/23/2022] Open
Abstract
Background Disruption of the lymphatic vasculature causes edema, inflammation, and end-tissue destruction. To assess the therapeutic efficacy of systemic anti-inflammatory therapy in this disease, we examined the impact of a nonsteroidal anti-inflammatory drug (NSAID), ketoprofen, and of a soluble TNF-α receptor (sTNF-R1) upon tumor necrosis factor (TNF)-α activity in a mouse model of acquired lymphedema. Methods and Findings Lymphedema was induced by microsurgical ablation of major lymphatic conduits in the murine tail. Untreated control mice with lymphedema developed significant edema and extensive histopathological inflammation compared to sham surgical controls. Short-term ketoprofen treatment reduced tail edema and normalized the histopathology while paradoxically increasing TNF-α gene expression and cytokine levels. Conversely, sTNF-R1 treatment increased tail volume, exacerbated the histopathology, and decreased TNF-α gene expression. Expression of vascular endothelial growth factor-C (VEGF-C), which stimulates lymphangiogenesis, closely correlated with TNF-α expression. Conclusions Ketoprofen therapy reduces experimental post-surgical lymphedema, yet direct TNF-α inhibition does not. Reducing inflammation while preserving TNF-α activity appears to optimize the repair response. It is possible that the observed favorable responses, at least in part, are mediated through enhanced VEGF-C signaling.
Collapse
Affiliation(s)
- Kenta Nakamura
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Kavita Radhakrishnan
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yat Man Wong
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Stanley G. Rockson
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
18
|
Mannosamine–biotin as a novel masking agent for coating IgG for immune response silencing and augmentation of antibody–antigen interaction. Vaccine 2009; 27:6869-76. [DOI: 10.1016/j.vaccine.2009.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Revised: 08/19/2009] [Accepted: 09/01/2009] [Indexed: 01/29/2023]
|
19
|
Abstract
The term PEGylation describes the modification of biological molecules by covalent conjugation with polyethylene glycol (PEG), a non-toxic, non-immunogenic polymer, and is used as a strategy to overcome disadvantages associated with some biopharmaceuticals. PEGylation changes the physical and chemical properties of the biomedical molecule, such as its conformation, electrostatic binding, and hydrophobicity, and results in an improvement in the pharmacokinetic behavior of the drug. In general, PEGylation improves drug solubility and decreases immunogenicity. PEGylation also increases drug stability and the retention time of the conjugates in blood, and reduces proteolysis and renal excretion, thereby allowing a reduced dosing frequency. In order to benefit from these favorable pharmacokinetic consequences, a variety of therapeutic proteins, peptides, and antibody fragments, as well as small molecule drugs, have been PEGylated. This paper reviews the chemical procedures and the conditions that have been used thus far to achieve PEGylation of biomedical molecules. It also discusses the importance of structure and size of PEGs, as well as the behavior of linear and branched PEGs. A number of properties of the PEG polymer--e.g. mass, number of linking chains, the molecular site of PEG attachment--have been shown to affect the biological activity and bioavailability of the PEGylated product. Releasable PEGs have been designed to slowly release the native protein from the conjugates into the blood, aiming at avoiding any loss of efficacy that may occur with stable covalent PEGylation. Since the first PEGylated drug was developed in the 1970s, PEGylation of therapeutic proteins has significantly improved the treatment of several chronic diseases, including hepatitis C, leukemia, severe combined immunodeficiency disease, rheumatoid arthritis, and Crohn disease. The most important PEGylated drugs, including pegademase bovine, pegaspargase, pegfilgrastim, interferons, pegvisomant, pegaptanib, certolizumab pegol, and some of the PEGylated products presently in an advanced stage of development, such as PEG-uricase and PEGylated hemoglobin, are reviewed. The adaptations and applications of PEGylation will undoubtedly prove useful for the treatment of many previously difficult-to-treat conditions.
Collapse
|
20
|
Graves SS, Ryoo HM, Sale G, Storb R, Cowan LA, Matsuda K. Pegylated TNF-alpha receptor does not prevent acute graft-versus-host disease in the dog leukocyte antigen-nonidentical unrelated canine model. Biol Blood Marrow Transplant 2007; 12:1198-200. [PMID: 17085313 DOI: 10.1016/j.bbmt.2006.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Accepted: 07/05/2006] [Indexed: 11/28/2022]
|
21
|
Taluja A, Youn YS, Bae YH. Novel approaches in microparticulate PLGA delivery systems encapsulating proteins. ACTA ACUST UNITED AC 2007. [DOI: 10.1039/b706939a] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
22
|
Abstract
Pegylation, generally described as the molecular attachment of polyethylene glycols (PEGs) with different molecular weights to active drug molecules or surface treatment of drug-bearing particles with PEGs, is one of the most promising and extensively studied strategies with the goal of improving the pharmacokinetic behavior of the therapeutic drugs. A variety of PEGs, both linear and branched, with different molecular weights have been exploited successfully for use in this procedure in the form of reactive PEG species. Both reversible and irreversible PEG-drug conjugates have been prepared with relative advantages/disadvantages. The main pharmacokinetic outcomes of pegylation are summarized as changes occurring in overall circulation life-span, tissue distribution pattern, and elimination pathway of the parent drug/particle. Based on these favorable pharmacokinetic consequences leading to desired pharmacodynamic outcomes, a variety of proteins/peptides as well as small molecule drugs have been pegylated and evaluated successfully. Also a number of corresponding products have been approved by the U.S. FDA for specific clinical indications and some others are underway. In this article, the chemistry, rationale, strategies, pharmacokinetic outcomes, and therapeutic possibilities of pegylated drugs are reviewed with pharmacokinetic aspects presented with more details.
Collapse
Affiliation(s)
- Mehrdad Hamidi
- Department of Pharmaceutics, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | | | |
Collapse
|
23
|
Lundberg P, Welander PV, Edwards CK, van Rooijen N, Cantin E. Tumor necrosis factor (TNF) protects resistant C57BL/6 mice against herpes simplex virus-induced encephalitis independently of signaling via TNF receptor 1 or 2. J Virol 2006; 81:1451-60. [PMID: 17108044 PMCID: PMC1797509 DOI: 10.1128/jvi.02243-06] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tumor necrosis factor (TNF) is a multifunctional cytokine that has a role in induction and regulation of host innate and adaptive immune responses. The importance of TNF antiviral mechanisms is reflected by the diverse strategies adopted by different viruses, particularly members of the herpesvirus family, to block TNF responses. TNF binds and signals through two receptors, Tnfrsf1a (TNF receptor 1 [TNFR1], or p55) and Tnfrsf1b (TNFR2, or p75). We report here that herpes simplex virus 1 (HSV-1) infection of TNF-/- mice on the resistant C57BL/6 genetic background results in significantly increased susceptibility (P < 0.0001, log rank test) to fatal HSV encephalitis (HSE) and prolonged persistence of elevated levels of virus in neural tissues. In contrast, although virus titers in neural tissues of p55-/- N13 mice were elevated to levels comparable to what was found for the TNF-/- mice, the p55-/- N13 mice were as resistant as control C57BL/6 mice (P > 0.05). The incidence of fatal HSE was significantly increased by in vivo neutralization of TNF using soluble TNFR1 (sTNFR1) or depletion of macrophages in C57BL/6 mice (P = 0.0038 and P = 0.0071, respectively). Strikingly, in vivo neutralization of TNF in HSV-1-infected p55-/- p75-/- mice by use of three independent approaches (treatment with soluble p55 receptor, anti-TNF monoclonal antibody, or in vivo small interfering RNA against TNF) resulted in significantly increased mortality rates (P = 0.005), comparable in magnitude to those for C57BL/6 mice treated with sTNFR1 (P = 0.0018). Overall, these results indicate that while TNF is required for resistance to fatal HSE, both p55 and p75 receptors are dispensable. Precisely how TNF mediates protection against HSV-1 mortality in p55-/- p75-/- mice remains to be determined.
Collapse
Affiliation(s)
- Patric Lundberg
- City of Hope Medical Center and Beckman Research Institute, Department of Virology, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | | | | | | | | |
Collapse
|
24
|
Williams RO, Inglis JJ, Simelyte E, Criado G, Sumariwalla PF. Analysing the effect of novel therapies on cytokine expression in experimental arthritis. Int J Exp Pathol 2006; 86:267-78. [PMID: 16191099 PMCID: PMC2517440 DOI: 10.1111/j.0959-9673.2005.00443.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Type II collagen-induced arthritis (CIA) is an animal model of rheumatoid arthritis that has been used extensively to address questions of disease pathogenesis and to validate novel therapeutic targets. Susceptibility to CIA is strongly associated with major histocompatibility complex class II genes, and the development of arthritis is accompanied by a robust T- and B-cell response to type II collagen. The main pathological features of CIA include proliferative synovitis with infiltration of inflammatory cells, pannus formation, cartilage degradation, erosion of bone and fibrosis. Pro-inflammatory cytokines, such as tumour necrosis factor alpha and interleukin-1beta, are expressed in the arthritic joints in both murine CIA and human rheumatoid arthritis, and blockade of these molecules results in amelioration of disease. Hence, there is a great deal of interest in the development of small-molecular-weight inhibitors of pro-inflammatory cytokines. There is also interest in the development and testing of drugs with the capacity to modulate the immune pathways involved in driving the inflammatory response in arthritis. For these reasons, there is a need to monitor the effect of novel treatments on cytokine expression in vivo. In this review, we outline the various techniques used to detect cytokines in experimental arthritis and describe how these techniques have been used to quantify changes in cytokine expression following therapeutic intervention.
Collapse
Affiliation(s)
- Richard O Williams
- Kennedy Institute of Rheumatology Division, Imperial College London, London, UK.
| | | | | | | | | |
Collapse
|
25
|
|
26
|
Edwards CK, Bendele AM, Reznikov LI, Fantuzzi G, Chlipala ES, Li L, Moldawer LL, Mountz JD, Li YYY, Dinarello CA. Soluble human p55 and p75 tumor necrosis factor receptors reverse spontaneous arthritis in transgenic mice expressing transmembrane tumor necrosis factor α. ACTA ACUST UNITED AC 2006; 54:2872-85. [PMID: 16947419 DOI: 10.1002/art.22077] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE The roles of the transmembrane and secreted forms of tumor necrosis factor alpha (TNFalpha) in rheumatoid arthritis (RA) remain unclear. Agents used to inhibit TNFalpha have shown varying efficacy in RA patients, suggesting that anti-TNFalpha agents possess dissimilar mechanisms of action, including the ability to neutralize transmembrane (tmTNFalpha) and secreted TNFalpha. In this study, TNFalpha-knockout (TNFalpha-KO) mice that were genetically altered to express elevated levels of tmTNFalpha were constructed to further understand the roles of the 17-kd secreted, trimeric, and 26-kd transmembrane forms of TNFalpha. METHODS A speed-congenic mating scheme was used to generate 3 unique strains of mice: 1) transgenic tmTgA86 mice overexpressing 26-kd tmTNFalpha and also secreting 17-kd trimeric TNFalpha (tmTNFalpha-transgenic), 2) TNFalpha-/- mice (TNFalpha-KO), and 3) transgenic mice overexpressing tmTNFalpha backcrossed to TNFalpha-KO mice (tmTNFalpha-transgenic/TNFalpha-KO). Mice were treated with phosphate buffered saline (as vehicle control), dexamethasone (as positive control), or modified recombinant human soluble TNF receptor (sTNFR) p55 or p75, and were assessed clinically and histopathologically for signs of inflammation and development of arthritis. RESULTS The tmTNFalpha-transgenic/TNFalpha-KO mice were born with crinkled tails and spinal deformities similar to those in ankylosing spondylitis. By 2-4 weeks, these mice developed symmetric inflammatory arthritis, characterized by tissue swelling, pannus formation, and bone deformities. The tmTNFalpha-transgenic mice also developed spontaneous-onset arthritis, but at a slower rate (100% incidence by 10-12 weeks). Clinical and histologic progression of arthritis in the tmTNFalpha-transgenic/TNFalpha-KO mice was reduced by treatment with dexamethasone or with the p55 or p75 sTNFR (69% and 63% reduction in total histologic score, respectively). CONCLUSION These data show that arthritis is sufficiently initiated and maintained in tmTNFalpha-transgenic/TNFalpha-KO mice, and that it can be neutralized by recombinant human p55 or p75 sTNFR, resulting in amelioration of the biologic and subsequent histologic destructive effects of tmTNFalpha.
Collapse
|
27
|
Takeuchi T, Amano K, Kameda H, Abe T. Anti-TNF Biological Agents in Rheumatoid Arthritis and Other Inflammatory Diseases. Allergol Int 2005. [DOI: 10.2332/allergolint.54.191] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
28
|
Schottelius AJG, Moldawer LL, Dinarello CA, Asadullah K, Sterry W, Edwards CK. Biology of tumor necrosis factor-alpha- implications for psoriasis. Exp Dermatol 2004; 13:193-222. [PMID: 15086336 DOI: 10.1111/j.0906-6705.2004.00205.x] [Citation(s) in RCA: 144] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Numerous recent investigations have pointed to a key role of the proinflammatory, pleiotropic cytokine tumor necrosis factor-alpha (TNF-alpha) in host defense and inflammatory processes. TNF overexpression has been found in lesional skin and in the circulation of psoriatic patients, and it was suggested that TNF-alpha is crucial in this and other immune diseases. Several approaches to inhibit TNF-alpha activity have been developed. These include three different neutralizing antibodies to TNF-alpha as well as three different soluble TNF-alpha receptors with characteristic properties designed to bind the 17-KDa soluble trimeric TNF-alpha and the 26-KDa membrane-bound form of TNF-alpha. Clinical trials have demonstrated significant antipsoriatic effects, and it is likely that blocking TNF-alpha will become an important therapeutic option. The data available from these trials contribute to further understanding of the disease by demonstrating the major role of TNF-alpha. An in-depth understanding of the regulation of TNF gene expression, protein production, receptor expression, and signaling pathways may lead to further, potentially important novel therapeutic strategies and antipsoriatic active small molecules, suitable for oral application in the future. Here we review the current knowledge of TNF biology, the approaches to inhibit TNF activity, and their clinical and immunological effects in psoriasis. In addition, the host-defense effects and chronic TNF-blocking activity are discussed.
Collapse
Affiliation(s)
- Arndt J G Schottelius
- Schering AG/Berlex Biosciences, Research Business Area Dermatology USA, Richmond, CA 94804-0099, USA
| | | | | | | | | | | |
Collapse
|
29
|
Campbell IK, Roberts LJ, Wicks IP. Molecular targets in immune-mediated diseases: the case of tumour necrosis factor and rheumatoid arthritis. Immunol Cell Biol 2003; 81:354-66. [PMID: 12969323 DOI: 10.1046/j.0818-9641.2003.01185.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Rheumatoid arthritis is a common autoimmune condition in which, for unknown reasons, synovial joints become the target of a sustained immune response. For many years, rheumatoid arthritis was in the 'too hard basket' in terms of understanding disease mechanisms and providing rational therapy. This has changed dramatically over the last 10 years and rheumatoid arthritis is now at the forefront of biotechnology. In this review, we outline one of the most exciting recent developments, namely antagonists of the cytokine TNF. The preclinical evaluation of TNF in animal models of rheumatoid arthritis, and subsequent clinical trials of TNF inhibitors in patients, provides insight into the 'bench to bedside' paradigm. We therefore briefly review rheumatoid arthritis, animal models of rheumatoid arthritis, the biology of TNF, the pivotal clinical trials of TNF antagonists and the emerging data on side-effects. Tumour necrosis factor inhibitors in rheumatoid arthritis represent the first attempt to achieve sustained blockade of a single cytokine in a human disease. Whilst this approach has been even more successful than might have been predicted, we suggest it is only the beginning of what has become a new therapeutic era.
Collapse
Affiliation(s)
- Ian K Campbell
- Reid Rheumatology Laboratory, Walter and Eliza Hall Institute of Medical Research, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
30
|
Abbas S, Zhang YH, Clohisy JC, Abu-Amer Y. Tumor necrosis factor-alpha inhibits pre-osteoblast differentiation through its type-1 receptor. Cytokine 2003; 22:33-41. [PMID: 12946103 DOI: 10.1016/s1043-4666(03)00106-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tumor necrosis factor-alpha (TNF) is a pro-inflammatory cytokine with a profound role in many skeletal diseases. The cytokine has been described as a mediator of bone loss in osteolysis and other inflammatory bone diseases. In addition to its known bone resorptive action, TNF reduces bone formation by inhibiting osteoblast differentiation. Using primary and transformed osteoblastic cells, we first document that TNF inhibits expression of alkaline phosphatase and matrix deposition, both considered markers of osteoblast differentiation. The effects are dose- and time-dependent. Core-binding factor A1 (cbfa1) is a transcription factor critical for osteoblast differentiation, and we show here that it is activated by the osteoblast differentiation agent, beta-glycerophosphate. Therefore, we investigated whether the inhibitory effects of TNF were associated with altered activity of this transcription factor. Using retardation assays, we show that TNF significantly inhibits cbfal activation by beta-glycerophosphate, manifested by reduced DNA-binding activity. Next, we turned to determine the signaling pathway by which TNF inhibits osteoblast differentiation. Utilizing animals lacking individual TNF receptors, we document that TNFr1 is required for transmitting the cytokine's inhibitory effect. In the absence of this receptor, TNF failed to impact all osteoblast differentiation markers tested. In summary, TNF blocks expression of osteoblast differentiation markers and inhibits beta-glycerophosphate-induced activation of the osteoblast differentiation factor cbfa1. Importantly, these effects are mediated via a mechanism requiring the TNF type-1 receptor.
Collapse
Affiliation(s)
- Sabiha Abbas
- Department of Orthopaedics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
31
|
Gardam MA, Keystone EC, Menzies R, Manners S, Skamene E, Long R, Vinh DC. Anti-tumour necrosis factor agents and tuberculosis risk: mechanisms of action and clinical management. THE LANCET. INFECTIOUS DISEASES 2003; 3:148-55. [PMID: 12614731 DOI: 10.1016/s1473-3099(03)00545-0] [Citation(s) in RCA: 500] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cases of active tuberculosis have been reported worldwide with the use of therapeutic agents that inhibit tumour necrosis factor (TNF) alpha. TNFalpha has a central role in mycobacterial infection and disease. Accordingly, progression of recently acquired tuberculosis infection or reactivation of remotely acquired infection should be expected with the use of anti-TNF agents. The available in-vitro and epidemiological evidence for the two currently approved agents, infliximab and etanercept, shows that the risk of development of active tuberculosis is greater with infliximab. Tuberculin skin testing (TST) should be undertaken before any significant immunosuppressive therapy including these agents, though the possibility of false-negative reactions in immunocompromised populations must be borne in mind. A positive TST should be followed by medical assessment and chest radiography, as well as by other tests judged appropriate by the physician to identify active disease. Active tuberculosis must be treated appropriately before initiation of treatment with an anti-TNF agent. Treatment of latent tuberculosis can be considered on an individual basis for TST-negative patients receiving anti-TNF agents when significant risk factors for infection are present.
Collapse
Affiliation(s)
- Michael A Gardam
- Infection Prevention and Control and Tuberculosis Clinic, Division of Infectious Disease, University Health Network, Ontario, Toronto, Canada.
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Protein and peptide drugs hold great promise as therapeutic agents. However, many are degraded by proteolytic enzymes, can be rapidly cleared by the kidneys, generate neutralizing antibodies and have a short circulating half-life. Pegylation, the process by which polyethylene glycol chains are attached to protein and peptide drugs, can overcome these and other shortcomings. By increasing the molecular mass of proteins and peptides and shielding them from proteolytic enzymes, pegylation improves pharmacokinetics. This article will review how PEGylation can result in drugs that are often more effective and safer, and which show improved patient convenience and compliance.
Collapse
Affiliation(s)
- J Milton Harris
- Nektar Therapeutics, 490 Discovery Drive, Huntsville, Alabama 35806, USA.
| | | |
Collapse
|
33
|
Abstract
Mirror-image allodynia is a mysterious phenomenon that occurs in association with many clinical pain syndromes. Allodynia refers to pain in response to light touch/pressure stimuli, which normally are perceived as innocuous. Mirror-image allodynia arises from the healthy body region contralateral to the actual site of trauma/inflammation. Virtually nothing is known about the mechanisms underlying such pain. A recently developed animal model of inflammatory neuropathy reliably produces mirror-image allodynia, thus allowing this pain phenomenon to be analyzed. In this sciatic inflammatory neuropathy (SIN) model, decreased response threshold to tactile stimuli (mechanical allodynia) develops in rats after microinjection of immune activators around one healthy sciatic nerve at mid-thigh level. Low level immune activation produces unilateral allodynia ipsilateral to the site of sciatic inflammation; more intense immune activation produces bilateral (ipsilateral + mirror image) allodynia. The present studies demonstrate that both ipsilateral and mirror-image SIN-induced allodynias are (1) reversed by intrathecal (peri-spinal) delivery of fluorocitrate, a glial metabolic inhibitor; (2) prevented and reversed by intrathecal CNI-1493, an inhibitor of p38 mitogen-activated kinases implicated in proinflammatory cytokine production and signaling; and (3) prevented or reversed by intrathecal proinflammatory cytokine antagonists specific for interleukin-1, tumor necrosis factor, or interleukin-6. Reversal of ipsilateral and mirror-image allodynias was rapid and complete even when SIN was maintained constantly for 2 weeks before proinflammatory cytokine antagonist administration. These results provide the first evidence that ipsilateral and mirror-image inflammatory neuropathy pain are created both acutely and chronically through glial and proinflammatory cytokine actions.
Collapse
|
34
|
Milligan ED, Twining C, Chacur M, Biedenkapp J, O'Connor K, Poole S, Tracey K, Martin D, Maier SF, Watkins LR. Spinal glia and proinflammatory cytokines mediate mirror-image neuropathic pain in rats. J Neurosci 2003; 23:1026-40. [PMID: 12574433 PMCID: PMC6741915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023] Open
Abstract
Mirror-image allodynia is a mysterious phenomenon that occurs in association with many clinical pain syndromes. Allodynia refers to pain in response to light touch/pressure stimuli, which normally are perceived as innocuous. Mirror-image allodynia arises from the healthy body region contralateral to the actual site of trauma/inflammation. Virtually nothing is known about the mechanisms underlying such pain. A recently developed animal model of inflammatory neuropathy reliably produces mirror-image allodynia, thus allowing this pain phenomenon to be analyzed. In this sciatic inflammatory neuropathy (SIN) model, decreased response threshold to tactile stimuli (mechanical allodynia) develops in rats after microinjection of immune activators around one healthy sciatic nerve at mid-thigh level. Low level immune activation produces unilateral allodynia ipsilateral to the site of sciatic inflammation; more intense immune activation produces bilateral (ipsilateral + mirror image) allodynia. The present studies demonstrate that both ipsilateral and mirror-image SIN-induced allodynias are (1) reversed by intrathecal (peri-spinal) delivery of fluorocitrate, a glial metabolic inhibitor; (2) prevented and reversed by intrathecal CNI-1493, an inhibitor of p38 mitogen-activated kinases implicated in proinflammatory cytokine production and signaling; and (3) prevented or reversed by intrathecal proinflammatory cytokine antagonists specific for interleukin-1, tumor necrosis factor, or interleukin-6. Reversal of ipsilateral and mirror-image allodynias was rapid and complete even when SIN was maintained constantly for 2 weeks before proinflammatory cytokine antagonist administration. These results provide the first evidence that ipsilateral and mirror-image inflammatory neuropathy pain are created both acutely and chronically through glial and proinflammatory cytokine actions.
Collapse
Affiliation(s)
- Erin D Milligan
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309-0345, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Bush KA, Walker JS, Frazier J, Kirkham BW. Effects of a PEGylated soluble TNF receptor type 1 (PEG sTNF-RI) on cytokine expression in adjuvant arthritis. Scand J Rheumatol 2002; 31:198-204. [PMID: 12369650 DOI: 10.1080/030097402320318378] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To investigate the effects of TNF blocking therapy on synovial immune activity in rat adjuvant arthritis (AA) by measuring mRNA expression of key macrophage and T cell cytokines during PEG sTNF-RI treatment (10mg/kg) on days 8, 10 and 12. METHODS Paw volume was assessed every 3-4 days. Ankles were removed for quantitative radiology and histology and synovial membrane removed to determine cytokine mRNA expression using semi-quantitative RT-PCR. T cells in joints were quantified by immunohistochemistry. RESULTS Paw volume was significantly decreased in rats treated with PEG STNF-RI from days 12 to 17. Histology scores and synovial T cell numbers were reduced on days 13 and 17 and radiology scores significantly reduced on day 13. Expression of synovial TNF, IFN-gamma, IL-17, IL-2 and IL-4 mRNA was unchanged in treated rats and TGF-beta expression was significantly increased at day 13. CONCLUSIONS PEG sTNF-RI attenuates AA and disease recurs after treatment ceases, similar to human rheumatoid arthritis. Continued TNF production and/or ongoing T cell activity, may explain the recrudescence of disease once treatment is stopped.
Collapse
Affiliation(s)
- K A Bush
- School of Physiology & Pharmacology, University of New South Wales, Sydney, Australia
| | | | | | | |
Collapse
|
36
|
Kerwin BA, Chang BS, Gegg CV, Gonnelli M, Li T, Strambini GB. Interactions between PEG and type I soluble tumor necrosis factor receptor: modulation by pH and by PEGylation at the N terminus. Protein Sci 2002; 11:1825-33. [PMID: 12070334 PMCID: PMC2373643 DOI: 10.1110/ps.0208102] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The effects of polyethylene glycol (PEG) on protein structure and the molecular details that regulate its association to polypeptides are largely unknown. These issues were addressed using type I soluble tumor necrosis factor receptor (sTNF-RI) as a model system. Changes in solution viscosity established that a truncated form of sTNF-RI bound free PEG in a pH-dependent manner. Above pH 5.3, the viscosity escalated as the pH increased, while no effect occurred below pH 5.0. Conjugation of 2 kD, 5 kD, or 20 kD PEG to the N terminus attenuated the viscosity at the higher pH values. Tryptophan phosphorescence spectroscopy correlated changes in the protein structure about Trp-107, at the C terminus, with the pH-dependent and PEGylation-dependent attenuation of the viscosity. The results indicate that specific interactions between PEG and the truncated form of sTNF-RI are elicited by an increased flexibility of the truncated protein combined perhaps with removal of steric or charge barriers. Covalently bound PEG at the N terminus reduced the protein affinity for the free polymer and induced a more rigid and polar configuration around Trp-107. Deprotonation of His-105, which is perpendicular to Trp-107, was integral to the binding mechanism producing a pH-dependent switching mechanism. These findings stress the importance of surface charge and structural plasticity in determining macromolecular binding affinities and demonstrate the ability of conjugated PEG to modify the localized surface structure in proteins away from the site of conjugation.
Collapse
Affiliation(s)
- Bruce A Kerwin
- Amgen, Inc., One Amgen Center Drive, Mail Stop 2-1-A, Thousand Oaks, CA 91320, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
The goal of this research was to determine whether the site-specific attachment of poly(ethylene glycol) to insulin could enhance the physical and pharmacological properties of insulin without negatively affecting its biological activity or immunological properties. Electrophilically activated derivatives of low-molecular-weight monomethoxypoly(ethylene glycol) (mPEG) were chemically coupled to insulin via its amino groups at positions phenylalanine-B1 or lysine-B29, with an amide bond being formed between the polymer and protein. The site-specific attachment of mPEG to insulin did not substantially alter insulin's secondary/tertiary structure, self-association behavior, or potency in vivo. However, mPEG attachment did significantly enhance insulin's resistance to aggregation. In addition, the pegylation of insulin almost completely eliminates the resultant conjugate's immunogenicity, allergenicity, and antigenicity. Finally, the conjugates were observed to remain in the systemic circulation for longer periods of time than unmodified insulin after subcutaneous administration.
Collapse
Affiliation(s)
- Kenneth D Hinds
- Department of Pharmaceutics and Pharmaceutical Chemistry/CCCD, University of Utah, 20 South 2030 East Rm. 201, Salt Lake City, UT 84112, USA
| | | |
Collapse
|
38
|
Abstract
Poly(ethylene glycol) (PEG) is a highly investigated polymer for the covalent modification of biological macromolecules and surfaces for many pharmaceutical and biotechnical applications. In the modification of biological macromolecules, peptides and proteins are of extreme importance. Reasons for PEGylation (i.e. the covalent attachment of PEG) of peptides and proteins are numerous and include shielding of antigenic and immunogenic epitopes, shielding receptor-mediated uptake by the reticuloendothelial system (RES), and preventing recognition and degradation by proteolytic enzymes. PEG conjugation also increases the apparent size of the polypeptide, thus reducing the renal filtration and altering biodistribution. An important aspect of PEGylation is the incorporation of various PEG functional groups that are used to attach the PEG to the peptide or protein. In this paper, we review PEG chemistry and methods of preparation with a particular focus on new (second-generation) PEG derivatives, reversible conjugation and PEG structures.
Collapse
Affiliation(s)
- M J Roberts
- Shearwater Corporation, 490 Discovery Drive, Huntsville, AL 35806, USA.
| | | | | |
Collapse
|
39
|
Abstract
Cytokines are a growing group of proteins that are responsible for the communication of cells of the immune system, hematopoietic cells, and other cell types. They play a dominant role in various diseases, particularly in promoting and perpetuating inflammation. Cytokine production is a reaction of the body to a pathologic state to restore homeostasis. In such cases, the therapeutic intervention should support the reaction of the body by giving the cytokine itself (agonistic therapeutics). In other cases, manifestation of a disease results from an overproduction of cytokines, making cytokine antagonists desirable therapeutic drugs. Furthermore, cytokines may be good candidates as cancer therapeutics, especially to support the restoration of blood cell populations after chemotherapy or radiation.
Collapse
Affiliation(s)
- Heidi Schooltink
- Department of Biochemistry, Christian-Albrechts-Universität Kiel, D-24098 Kiel, Germany
| | | |
Collapse
|
40
|
Abstract
The aetiology of systemic, autoimmune, chronic inflammatory diseases--such as rheumatoid arthritis--is not known, and their pathogenesis is complex and multifactorial. However, progress in the characterization of intercellular mediators--proteins that are now known as cytokines--has led to the realization that one cytokine, tumour-necrosis factor (TNF; previously known as TNF-alpha), has an important role in the pathogenesis of rheumatoid arthritis. This discovery heralded a new era of targeted and highly effective therapeutics for rheumatoid arthritis and, subsequently, other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Marc Feldmann
- Kennedy Institute, Rheumatology Division, Faculty of Medicine, Imperial College School of Medicine, London, UK.
| |
Collapse
|
41
|
Zaccone P, Fehérvári Z, Blanchard L, Nicoletti F, Edwards CK, Cooke A. Autoimmune thyroid disease induced by thyroglobulin and lipopolysaccharide is inhibited by soluble TNF receptor type I. Eur J Immunol 2002; 32:1021-8. [PMID: 11920568 DOI: 10.1002/1521-4141(200204)32:4<1021::aid-immu1021>3.0.co;2-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Experimental autoimmune thyroiditis (EAT) is inducible in mice by immunization with thyroglobulin and adjuvant. Previous studies have shown that EAT is an autoimmune Th1-mediated disease but its characteristics differ with the adjuvant. Granulomatous lesions with marked follicular disruption develop following administration of thyroglobulin (Tg) and complete Freund's adjuvant (CFA) whereas when lipopolysaccharide (LPS) is used as the adjuvant only focal infiltrates of mononuclear cells are observed. The pro-inflammatory cytokine, TNF-alpha, is associated with Th1 autoimmune-mediated conditions. Cytokine antagonists have been used as potential therapeutic agents in several experimental autoimmune models. Soluble cytokine receptors belong to this category and may naturally be shed from cell membranes to inhibit cytokine activity. We show that the administration of the soluble TNF receptor type I (sTNFR I) in the induction of EAT has very different effects on the two models of induced autoimmune thyroiditis. sTNFR I treatment inhibits the induction of EAT only when mouse Tg is given with LPS not with CFA, suggesting an important difference in the pathogenic processes.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Animals
- Antigens, CD/genetics
- Antigens, CD/pharmacology
- Antigens, CD/physiology
- Drug Administration Schedule
- Female
- Freund's Adjuvant
- Immunization
- Lipopolysaccharides/administration & dosage
- Lipopolysaccharides/immunology
- Lipopolysaccharides/toxicity
- Macrophages, Peritoneal/metabolism
- Male
- Mice
- Mice, Inbred CBA
- Mice, Inbred NOD
- Models, Animal
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Type I
- Recombinant Fusion Proteins/pharmacology
- Solubility
- Th1 Cells/immunology
- Thyroglobulin/administration & dosage
- Thyroglobulin/immunology
- Thyroglobulin/toxicity
- Thyroiditis, Autoimmune/drug therapy
- Thyroiditis, Autoimmune/immunology
- Thyroiditis, Autoimmune/physiopathology
- Thyroiditis, Autoimmune/prevention & control
- Tumor Necrosis Factor-alpha/physiology
Collapse
Affiliation(s)
- Paola Zaccone
- Department of Pathology, Immunology Division, Cambridge University, Cambridge, GB
| | | | | | | | | | | |
Collapse
|
42
|
Bush KA, Kirkham BW, Walker JS. The in vivo effects of tumour necrosis factor blockade on the early cell mediated immune events and syndrome expression in rat adjuvant arthritis. Clin Exp Immunol 2002; 127:423-9. [PMID: 11966757 PMCID: PMC1906318 DOI: 10.1046/j.1365-2249.2002.01742.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Anti-TNF therapy is effective in rheumatoid arthritis (RA); however, its mechanisms of action are incompletely understood. T cell-driven mechanisms are thought to play an important role in RA and the effects of TNF blockade on these mechanisms are unclear. Adjuvant arthritis (AA) is a T cell dependent model of inflammatory arthritis. The aims of this study were to investigate the effects of TNF blockade on in vivo T cell cytokine expression and to clarify the role of TNF in the inguinal lymph nodes (ILN) in early arthritis. AA was induced in male DA rats. Rats received either 3 mg/kg or 10 mg/kg PEG sTNF-RI at days 0, 2 and 4 postinduction or 10 mg/kg anti-TNF antibody on day of arthritis induction. Control rats received either saline or normal sheep serum. Paw volume was assessed every 3-4 days. Rats were sacrificed on days 0, 6, 13 and 21 postinduction. Ankles were removed for quantitative radiology and histology. Synovium and ILN were removed for cell culture and to determine mRNA expression of cytokines using semiquantitative RT-PCR. TNF and IFN-gamma protein production was measured using a bioassay and an ELISA. TNF blockade did not suppress mRNA expression of T cell cytokines in the ILN of rats in the early phase of AA, suggesting ongoing T cell activity. TNF protein production by ILN cells in culture was reduced in PEG sTNF-RI treated rats, although mRNA expression was increased in the ILN prior to culture. Early administration of PEG sTNF-RI did not attenuate AA, in contrast to an anti-TNF antibody, which suppressed disease. A shorter half-life for the PEG sTNF-RI compared with the anti-TNF antibody or the development of anti-PEG sTNF-RI antibodies may account for these results.
Collapse
Affiliation(s)
- K A Bush
- School of Physiology & Pharmacology, University of New South Wales, NSW, Australia
| | | | | |
Collapse
|
43
|
Abstract
Inflammation in rheumatoid arthritis (RA) is associated with an imbalance between pro- and anti-inflammatory factors, which leads to a persistent chronic inflammatory state in the joint. Molecular studies of the physiology of the inflammatory response have identified a hierarchy of cytokine activities. The identification of this hierarchy has provided new potential therapeutic targets for the treatment of RA. At present the majority of new therapeutic agents have been developed to neutralise the activity of tumour necrosis factor-alpha (TNF alpha), a cytokine at the top of the inflammatory cascade. These agents consist of recombinant proteins that bind and neutralise TNF alpha, and they are effective in the treatment of inflammation in RA. In this review we discuss the rationale behind targeting TNF alpha, the various recombinant proteins that have been used, their clinical effectiveness, the possible adverse effects of these agents and the development of new chemical inhibitors of TNF alpha synthesis.
Collapse
Affiliation(s)
- G J Pearce
- Department of Rheumatology & Bone and Joint Research Unit, St Bartholowmew's and the Royal London Hospital School of Medicine and Dentistry, Queen Mary and Westfield College, London, England
| | | |
Collapse
|
44
|
Zhang HG, Wang Y, Xie JF, Liang X, Liu D, Yang P, Hsu HC, Ray RB, Mountz JD. Regulation of tumor necrosis factor alpha-mediated apoptosis of rheumatoid arthritis synovial fibroblasts by the protein kinase Akt. ARTHRITIS AND RHEUMATISM 2001; 44:1555-67. [PMID: 11465707 DOI: 10.1002/1529-0131(200107)44:7<1555::aid-art279>3.0.co;2-m] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To determine if tumor necrosis factor alpha (TNFalpha)-driven proliferation of rheumatoid arthritis synovial fibroblasts (RASF) is associated with up-regulation of the activity of serine/threonine kinase B/Akt and with survival of RASF. METHODS Staining of phosphorylated Akt was done using anti-phosphorylated Thr308 Akt antibody. Levels of phosphorylated Akt were analyzed by Western blot and Akt activity was analyzed using a kinase assay. TUNEL staining was used to analyze the cytotoxicity of TNFalpha treatment or TNFalpha combined with either the Akt activity inhibitor wortmannin, an adenovirus expressing dominant-negative mutant (AdAkt-DN), or an adenovirus expressing phosphatase and tensin homolog deleted on chromosome 10 (AdPTEN). RESULTS The levels of phosphorylated Akt were higher in RASF than in osteoarthritis synovial fibroblasts (OASF), as demonstrated by immunohistochemical staining, immunoblot analysis, and an Akt kinase assay. The levels of phosphorylated Akt and Akt kinase activity were increased by stimulation of primary RASF with TNFalpha (10 ng/ml). Treatment of RASF with the phosphatidylinositol 3-kinase inhibitor wortmannin (50 nM) plus TNFalpha resulted in apoptosis of 60 +/- 8% (mean +/- SEM) of RASF within 24 hours. This proapoptosis effect was specific for Akt, since equivalent levels of apoptosis were observed upon TNFalpha treatment of RASF transfected with AdAkt-DN and with AdPTEN, which opposes the action of Akt. CONCLUSION These results indicate that phosphorylated Akt acts as a survival signal in RASF and contributes to the stimulatory effect of TNFalpha on these cells by inhibiting the apoptosis response. This effect was not observed in OASF and may reflect the pathophysiologic changes associated with the proliferating synovium in rheumatoid arthritis.
Collapse
Affiliation(s)
- H G Zhang
- The University of Alabama at Birmingham, 35294, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Diwan M, Park TG. Pegylation enhances protein stability during encapsulation in PLGA microspheres. J Control Release 2001; 73:233-44. [PMID: 11516501 DOI: 10.1016/s0168-3659(01)00292-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
During encapsulation of proteins in biodegradable microspheres, a significant amount of the protein reportedly undergoes denaturation to form irreversible insoluble aggregates. Incomplete in vitro release of proteins from the microspheres is a common observation. An attempt was made to overcome this problem by pegylation of the protein to be encapsulated. Lysozyme, a model protein, was conjugated with methoxy polyethylene glycol (mPEG, MW 5000). The conjugate was characterized by SDS-PAGE, SE-HPLC, and MALDI-TOF mass spectroscopy. The pegylated lysozyme (Lys-mPEG) consisted of different isomers of mono-, di- and tri-pegylated with about 15% as native lysozyme. The specific activity of the protein was retained after pegylation (101.3+/-10.4%). The microsphere encapsulation process was simulated for pegylated and native lysozyme. Pegylated lysozyme exhibited much better stability than native lysozyme against exposure to organic solvent (dichloromethane), homogenization, and showed reduced adsorption onto the surface of blank PLGA microspheres. Release profiles of the two proteins from microspheres were very different. For native lysozyme, it was high initial release (about 50%) followed by a nearly no release (about 10% in 50 days). In contrast, Lys-mPEG conjugate showed a triphasic and near complete release over 83 days. This study shows that the pegylation of protein can provide substantial protection against the destabilization of protein during encapsulation.
Collapse
Affiliation(s)
- M Diwan
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 305-701, Taejon, South Korea
| | | |
Collapse
|
46
|
Sweitzer S, Martin D, DeLeo JA. Intrathecal interleukin-1 receptor antagonist in combination with soluble tumor necrosis factor receptor exhibits an anti-allodynic action in a rat model of neuropathic pain. Neuroscience 2001; 103:529-39. [PMID: 11246166 DOI: 10.1016/s0306-4522(00)00574-1] [Citation(s) in RCA: 276] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The expression of interleukin-1beta and tumor necrosis factor has previously been shown to be up-regulated in the spinal cord of several rat mononeuropathy models. This present study was undertaken to determine whether blocking the action of central interleukin-1beta and tumor necrosis factor attenuates mechanical allodynia in a gender-specific manner in a rodent L5 spinal nerve transection model of neuropathic pain, and whether this inhibition occurs via down-regulation of the central cytokine cascade or blockade of glial activation. Interleukin-1 receptor antagonist or soluble tumor necrosis factor receptor was administered intrathecally via lumbar puncture to male Holtzman rats in a preventative pain strategy, in which therapy was initiated 1h prior to surgery. Administration of soluble tumor necrosis factor receptor attenuated mechanical allodynia, while interleukin-1 receptor antagonist alone was unable to decrease allodynia. Interleukin-1 receptor antagonist in combination with soluble tumor necrosis factor receptor, administered to both male and female rats in a preventative pain strategy, significantly reduced mechanical allodynia in a dose-dependent manner (P<0.01). The magnitude of attenuation in allodynia was similar in both males and females. Immunohistochemistry on L5 spinal cord revealed similar astrocytic and microglial activation regardless of treatment. At days 3 and 7 post-transection, animals receiving daily interleukin-1 receptor antagonist in combination with soluble tumor necrosis factor receptor exhibited significantly less interleukin-6, but not interleukin-1beta, in the L5 spinal cord compared to vehicle-treated animals. In an existing pain paradigm, in which treatment was initiated on day 7 post-transection, interleukin-1 receptor antagonist in combination with soluble tumor necrosis factor receptor attenuated mechanical allodynia (P<0.05) in male rats. These findings further support a role for central interleukin-1beta and tumor necrosis factor in the development and maintenance of neuropathic pain through induction of a proinflammatory cytokine cascade.
Collapse
Affiliation(s)
- S Sweitzer
- Department of Pharmacology and Toxicology, Hinman Box 7650, Dartmouth College, Hanover, NH 03755, USA.
| | | | | |
Collapse
|
47
|
Kozlowski A, Harris JM. Improvements in protein PEGylation: pegylated interferons for treatment of hepatitis C. J Control Release 2001; 72:217-24. [PMID: 11390000 DOI: 10.1016/s0168-3659(01)00277-2] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Poly(ethyleneglycol) or PEG has proven to be of great value for a range of biomedical applications. A review the properties of PEG that lead to these applications is reported. Emphasis is placed on pharmaceutical uses of PEG--proteins, with specific discussion of the attributes of PEGylated alpha-interferon for treatment of hepatitis C. In this latter case the choice of PEG reagent is critical to the properties of the drug, and therefore a brief presentation of PEG reagents for protein PEGylation will be given. PEGylation chemistries can be divided into first- and second-generation approaches. The first-generation chemistries are generally restricted to low-molecular-weight methoxy-PEGs because of the problem of diol contamination and resulting difunctional reagents. Problems with weak linkages and side reactions are also encountered. Second-generation PEGylation reagents avoid weak linkages and side reactions. Also they can be purified to remove diol contaminants, and as a consequence, high-molecular-weight PEGs can be used. These relatively simple chemical advances have given new vigor to PEGylation as a technology. The benefits of using high-molecular-weight, second-generation PEG reagents are demonstrated by using PEG--alpha-interferon as an example. In this case it is observed that a greatly improved drug is provided for treatment of hepatitis C.
Collapse
Affiliation(s)
- A Kozlowski
- Shearwater Corporation, 1112 Church St., Huntsville, AL 35801, USA
| | | |
Collapse
|
48
|
Reneses S, Pestana L. [Systematic review of clinical trials on the treatment of rheumatoid arthritis with tumour necrosis factor alpha inhibitors]. Med Clin (Barc) 2001; 116:620-8. [PMID: 11412650 DOI: 10.1016/s0025-7753(01)71925-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- S Reneses
- Servicio de Reumatología, Hospital Universitario Virgen del Rocío, Sevilla
| | | |
Collapse
|
49
|
Keystone EC. Tumor necrosis factor-alpha blockade in the treatment of rheumatoid arthritis. Rheum Dis Clin North Am 2001; 27:427-43. [PMID: 11396102 DOI: 10.1016/s0889-857x(05)70211-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The use of TNF alpha antagonists in RA has been extremely instructive. They have taught us that selective targeting of a pathogenic element can provide substantial clinical benefit. They have reinforced the concept of TNF alpha as a pivotal cytokine in the pathogenesis of RA. Pharmacodynamic studies of TNF alpha antagonists have further clarified the pathogenic processes involved in the disease. TNF alpha antagonists have set a new therapeutic standard for RA. Indeed, they are one of the most important advances in the history of the treatment of the disorder. If clinical efficacy is sustained and the safety profile remains benign over the long term, TNF alpha antagonists may replace MTX as the gold standard and become the agent of choice for combination therapy in RA. Further studies are needed to clarify their ultimate position in the therapeutic algorithm.
Collapse
Affiliation(s)
- E C Keystone
- Centre for Advanced Therapeutics in Arthritis, Mount Sinai Hospital, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Vázquez-Torres A, Fantuzzi G, Edwards CK, Dinarello CA, Fang FC. Defective localization of the NADPH phagocyte oxidase to Salmonella-containing phagosomes in tumor necrosis factor p55 receptor-deficient macrophages. Proc Natl Acad Sci U S A 2001; 98:2561-5. [PMID: 11226278 PMCID: PMC30177 DOI: 10.1073/pnas.041618998] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Tumor necrosis factor receptor (TNFR) p55-knockout (KO) mice are susceptible profoundly to Salmonella infection. One day after peritoneal inoculation, TNFR-KO mice harbor 1,000-fold more bacteria in liver and spleen than wild-type mice despite the formation of well organized granulomas. Macrophages from TNFR-KO mice produce abundant quantities of reactive oxygen and nitrogen species in response to Salmonella but nevertheless exhibit poor bactericidal activity. Treatment with IFN-gamma enhances killing by wild-type macrophages but does not restore the killing defect of TNFR-KO cells. Bactericidal activity of macrophages can be abrogated by a deletion in the gene encoding TNFalpha but not by saturating concentrations of TNF-soluble receptor, suggesting that intracellular TNFalpha can regulate killing of Salmonella by macrophages. Peritoneal macrophages from TNFR-KO mice fail to localize NADPH oxidase-containing vesicles to Salmonella-containing vacuoles. A TNFR-KO mutation substantially restores virulence to an attenuated mutant bacterial strain lacking the type III secretory system encoded by Salmonella pathogenicity island 2 (SPI2), suggesting that TNFalpha and SPI2 have opposing actions on a common pathway of vesicular trafficking. TNFalpha-TNFRp55 signaling plays a critical role in the immediate innate immune response to an intracellular pathogen by optimizing the delivery of toxic reactive oxygen species to the phagosome.
Collapse
Affiliation(s)
- A Vázquez-Torres
- Department of Medicine, University of Colorado Health Sciences Center, 4200 East 9th Avenue, Denver, CO 80262, USA
| | | | | | | | | |
Collapse
|