The usefulness and the limitations of animal models in identifying targets for therapy in arthritis

The usefulness and the limitations of animal models in identifying targets for therapy in arthritis

Best Practice & Research Clinical Rheumatology Vol. 18, No. 1, pp. 47–58, 2004 doi:10.1016/j.berh.2003.09.007 available online at http://www.sciencedi...

130KB Sizes 1 Downloads 38 Views

Best Practice & Research Clinical Rheumatology Vol. 18, No. 1, pp. 47–58, 2004 doi:10.1016/j.berh.2003.09.007 available online at http://www.sciencedirect.com

5 The usefulness and the limitations of animal models in identifying targets for therapy in arthritis Paul H. Wooley*

PhD

Professor of Orthopaedic Surgery Department of Orthopaedic Surgery, Wayne State University School of Medicine, 1 South, Hutzel Hospital, 4707 St. Antonie Blvd, Detroit, MI 48201, USA

Animal models have played a critical role in the history of modern drug development for rheumatoid arthritis (RA). In this chapter I examine the contributions of animal models in arthritis therapy from adjuvant arthritis and COX-1 inhibitors to transgenic mice and biological response modifiers. Advances in knowledge of the mechanisms of connective tissue disease are frequently derived from the study of animal models, and these findings frequently identify therapeutic targets that are subsequently evaluated in animal models. Hence a critical relationship between insights into the pathology of arthritis and the development of novel therapeutic approaches exists around the study of animal models of arthritis. In particular, we examine how the study of collagen-induced arthritis in rodents led to pioneering work in cytokine inhibitors for the successful therapy of RA. Key words: rheumatoid arthritis; animal models; biological response modifiers; drugs; immunotherapy.

The last 25 years of arthritis research have seen marked advances in our understanding of the basic pathology of connective tissue disease. This may be attributed in large part to the development of molecular techniques to address basic disease mechanisms, but it is not accidental that the initiation of this period coincides with the discovery of type II collagen-induced arthritis (CIA).1 This observation does not imply that highly significant findings were not discovered using adjuvant arthritis or antigen-induced arthritis (AIA), but merely contends that the coincidence of an attractive model with significant advances in technology inevitably will drive the state of the art in any research field. In more recent years, transgenic technology has risen to the forefront with the discovery that genetic manipulation at a single locus can generate useful models of experimental arthritis. Others may well look back in another decade * Tel.: þ 1-313-745-6828; Fax: þ1-313-993-0857. E-mail address: [email protected] (P.H. Wooley). 1521-6942/$ - see front matter Q 2004 Elsevier Ltd. All rights reserved.

48 P. H. Wooley

and decide that the ‘induced’ arthritides simply paved the way for a golden age of transgenic models, but one must also ponder whether sufficient rheumatoid patients will abandon their cytokine inhibition therapy to provide clinical testing for the next generation of drugs. In this chapter I review the progress of therapeutic advances in arthritis and the relationship to the advances in our knowledge of connective tissue disease derived from the study of animal models of arthritis.

EARLY MODELS OF ARTHRITIS Adjuvant arthritis became the benchmark for the evaluation of non-steroidal antiinflammatory drugs (NSAIDs, particularly COX-1 inhibitors) during the early 1960s.2 Although a highly useful model for the development of this drug modality, its precision in this employ also reflects the limitation of the model. COX-1 inhibitors can essentially cure adjuvant arthritis, but in human disease the anti-inflammatory and analgesic effects have far less influence upon the progression of erosive rheumatoid disease. This demonstrates a major tenet of drug development in animal models– models can represent only a fragment of the complex pathology of a disease such as rheumatoid arthritis (RA). This is further complicated by an appreciation that the human disease is a syndrome, and may exist in patients owing to a spectrum of environmental insults and immunogenetic regulation. Inbred animals subjected to a specific arthritogenic stimulus cannot be expected to represent the complex modality of RA. It is up to the investigator to discern whether the model represents a relevant aspect of the human pathology and to draw appropriate conclusions. The strength of models is drawn from the absence of the confounding factors that may ultimately explain the limited efficacy of a therapy that can occur when applied to the human condition. Another positive feature of adjuvant arthritis may be the ‘non-specific’ nature of the immune stimulation that results in joint disease. The role of the mineral oil and the mycobacterial components of complete Freund’s adjuvant (CFA) in the elicitation of joint disease still remains unclear, which is reminiscent of the unknown aetiology of RA. Despite the observation that both incomplete Freund’s adjuvant (IFA) and the mineral oil pristane may also induce arthritis in both rats and mice3,4, the arthritogenic process appears to depend upon either exogenous or autologous antigens such as heat-shock proteins (Hsp).5 Autoimmune T and B cell populations may arise due to the systemic overstimulation of macrophages by the adjuvant activation process, and give rise to joint infiltration and the production of a spectrum of autoantibodies against cartilage antigens and Hsp. In addition, macrophage-derived cytokines act directly upon synovial cells and chondrocytes, resulting in synovial hypertrophy, abnormal expression of MHC class II antigens and adhesion molecules, and atypical production of matrix components. However, another limit of the adjuvant arthritis in modelling RA may be the rapid disease onset (16 – 19 days), which is scarcely representative of the insidious onset and chronic nature of rheumatoid disease. Nevertheless, a rapid onset of disease is viewed as a major advantage in the pharmaceutical industry where cost considerations may take precedence over the scientific accuracy of a model. Another early model, AIA also contributed to the development of therapeutic strategies. AIA is elicited in rabbits and most rodent species by the injection of a soluble protein antigen (typically methylated bovine serum albumin) into the knee joint of an animal previously hypersensitized by immunization with the same antigen in Freund’s Complete Adjuvant (FCA). The simplicity might suggest that AIA is the consequence of any intra-articular immune response, but this characterization appears to be an

Animal models and arthritis therapy 49

oversimplification. The antigen must persist within the synovial joint, and the immunogenetic regulation of the response to antigen appears to contribute to arthritis susceptibility.6 The model is limited to an arthritis confined to the injected joint7, although the histological picture of a predominantly T-lymphocyte (mostly CD4þ) joint infiltrate, with B lymphocytes, mast cells and macrophages does provide a reasonably accurate model of RA.

COLLAGEN-INDUCED ARTHRITIS It did not take long from the initial description of CIA in rats1 to the first appearance of papers documenting the performance of the model using established rheumatoid drug treatments.8,9 CIA is only weakly responsive to NSAIDs or methotrexate, and is essentially unaffected by gold salts, chloroquine, colchicine or levamisole. The observation of poor NSAID activity in CIA was viewed by many as a distinct advantage over the adjuvant arthritis model. The nature of CIA, with its MHC-restricted autoimmune reaction to a native joint protein, represents a logical disease mechanism which closely resembles several aspects of RA. Indeed, reactivity to type II collagen appears to be quite prevalent in RA patients10 – 14, although the significance of this response in the disease pathology has never been firmly established. The CIA model provided several potential therapeutic targets, and the association of MHC linkage in both CIA and RA remains an intriguing and underexplored approach to this day. One working hypothesis for the immunopathology of RA remains the idea that joint antigens are presented in context of HLA-DR4 to autoreactive T cells, which trigger the autoimmune connective tissue disease. Thus, interference with DR4-restricted antigen presentation should block disease development. Initial studies using CIA supported this approach, with antibodies to H-2q surface antigens resulting in a marked protection against disease induction. Unfortunately, the therapeutic application of anti-Ia antibodies was less effective, probably due to the established presence of high levels of autoantibody and activated T cells within the joint. However, this target might still be appropriate for RA which (unlike rodent CIA) is characterized by exacerbations and remissions, and might be susceptible to the prevention of renewed immune activation as the disease progresses through cycles. The concern that MHC interference would be strongly immunosuppressive may be countered by the observation that most patients are heterozygous for class II MHC, and homozygous RA patients could be easily identified and exempted from treatment. Unpublished rumours have circulated describing fatal immunotoxic effects of MHC blockade in primates, and it would be useful to know whether this complication is valid. This approach could circumvent the requirement for strict knowledge of the nature of the antigen, or the T cell subset involved in the MHC-restricted arthritogenic response, but the development of MHC blocking peptides (see below) will probably render the development of this therapy unlikely. Immunotherapy for autoimmune arthritis became focussed upon the T cell with the discovery that cyclosporin prevented the onset of arthritis and slowed the progression of established disease in CIA.15 This phenomenon was confirmed in AIA, pristaneinduced arthritis16, proteoglycan-induced arthritis17 and streptococcal-cell-wallinduced arthritis18, and ultimately led to trials of anti-CD4 antibody therapy in RA. The clinical findings were less than spectacular, but this does not (in my opinion) reflect upon a failure of our model systems. Most rodent T cell depletion studies were not overly concerned with large reductions in circulating CD4 T cell levels, and many

50 P. H. Wooley

experients achieved levels below 5% of normal cell numbers. This aggressive approach was considered risky for human therapy, although there were no reports of problems with opportunistic infections or malignancy in the animal models. Combined with weakly therapeutic doses, the initial systemic delivery of anti-CD4 (rather than direct injection into the joint) made the outcome of clinical trials19 – 21 unsurprising to many immunologists, and the lack of high efficacy of anti-CD4 therapy in RA probably reflects a failure to target autoimmune T cells effectively (particularly synovial resident T cells) rather than the failure of the conceptual approach. Recent investigations using a primatized non-depleting anti-CD4 injected into the knees of RA patients indicated efficacy using MRI, arthroscopic, and histological imaging22, and CD4 T cell targeted immunotherapy may yet prove of benefit in RA.23 Targeting specific T cell subsets, based upon observations of skewing of T cell populations in affected joints24 has produced mixed results in collagen arthritis. Immunization against Vb peptides using BUB mice elicited antibodies reactive with the self-TCR and prevented the induction of CIA by eliminating or down-regulating pathogenic T cells25, and antibodies to specific Vb subsets have also been successful in CIA26,27, although not all researchers have experienced success with this approach.28,29 Although there is some evidence of T cell subset skewing in patients with RA30 – 32 it appears unlikely that there is sufficient restriction in T cell receptor usage in the disease to make this approach to immunotherapy viable. The most obvious success for the CIA model in therapeutic development can be attributed to the evaluation of cytokine inhibitors. Protocols for inhibiting the activity of IL-1b using IL-1 receptor antagonist protein (IRAP or IL-1ra)33 or antibodies to IL-1b34 have proved successful in CIA, and strategies for inhibiting TNF-a activity using soluble TNF receptor, recombinant TNF receptor:human Fc fusion protein, or antibodies to TNF have demonstrated potent anti-arthritic activity in CIA.35 – 37 These pre-clinical findings contributed to the successful development of anti-cytokine therapy for RA38 – 40, arguably the most significant advance in treatment for this disease since the discovery of steroids. Beyond the inhibition of inflammatory cytokines, CIA has indicated potential effects for other immune regulatory cytokines. It has been suggested that transforming growth factor (TGF)-b might be a suitable candidate36,41, but reports that TGF-b exerts an anti-arthritic effect in CIA are controversial, with some researchers finding that TGF-b may accelerate disease.42 It may prove problematic to utilize TGF-b effectively in RA because the effects of this cytokine on chondrocytes, as well as on immune cells, may make the outcome unpredictable. Antibodies to IL-6 have been shown to be efficacious in primate CIA43, and human trials using this approach are reported to show early success.44,45 Because the delivery of a cytokine inhibitor (which typically has a remarkably short plasma half-life) to the site of joint inflammation raises several practical problems, researchers have employed gene therapy techniques to examine the potential of this novel approach in arthritis.46 Systemic delivery of a TNFaR gene via adenoviral vectors was shown to reduce the severity of collagen-arthritis in rats, but was limited in intraarticular use due to the pro-inflammatory nature of the vector.47 Systemic delivery of chimeric human p55 TNFR-IgG fusion protein via adenovirus vector to mouse CIA had a short-term beneficial effect48, but again the outcome may have been compromised. It has been suggested that IL-4 might be an appropriate therapeutic for use in RA49 because this cytokine may exert an influence on the Th1/Th2 ratio within the rheumatoid joint. IL-4 introduced via an adenovirus vector into joints of mice with CIA caused over-expression of IL-4 in the mouse knee joint50, accompanied by enhanced onset and aggravation of the synovial inflammation. However, the histological analysis

Animal models and arthritis therapy 51

suggested prevention of chondrocyte death and cartilage erosion, with enhanced proteoglycan synthesis in articular cartilage. There have been several recent reports of success in CIA using gene therapeutic techniques to deliver IL-451 – 53, and it appears that this approach may have promise for the treatment of RA. The injection of viral IL10 via an adenoviral vector carrying the IL-10 gene into paw suppressed the development of collagen arthritis and reduced the progression of disease to noninjected paws, despite the absence of detectable levels of viral IL-10 in serum. Antiheterologous CII antibodies were unaffected, but IL-10 suppressed autoantibodies to murine type II collagen.54 Recently, vectors containing the murine IL-18 binding protein isoform c gene55, IL-1056, and IL-1ra57 have been demonstrated to be effective following intra-articular injection using CIA. Success using IRAP-mediated gene therapy in several models of arthritis58 has led to the first human trials of gene therapy in RA59, but it remains to be determined whether this approach represents the new future for arthritis therapy. Gene therapeutic approaches are now extending beyond the delivery of anti-inflammatory cytokines and inhibitors. A novel antigen-specific T-cell-mediated gene therapy has been demonstrated60 using T cell hybridomas specific for type II collagen, transduced to express an IL-12 antagonist and injected into collagenimmunized DBA/1 mice. The cell transfer was without effect on systemic T and B cells responses to CII, although antigen-elicited gIFN production was decreased while IL-4 responses in vitro were augmented. Collagen-reactive T cells migrated to inflammed joints, with a subsequent reduction or blockade of arthritis. Because these results could be reproduced in non-transformed CD4þ CII-reactive T cells it is possible that this approach could be applied to clinical trials. Collagen arthritis has also provided powerful evidence for a role of tolerance induction in the treatment of arthritis. Modulating the autoimmune response by the induction of tolerance might represent a key to the therapy of rheumatoid disease, and administration of type II collagen prior to the induction of CIA has proved to be effective in both rats and mice.61 Tolerant rats exhibited lower anti-collagen antibodies, as well as lower T cell proliferation responses to collagen in vitro. Oral administration of CII appears to alter the subsequent immune response to the arthritogenic challenge via an antigen-driven active suppression mechanism that affects both T cells and B-cells (rather than inducing clonal deletion or anergy) and may be implemented by the regulatory cytokines IL-4, IL-10 and TGF-b. Studies on the induction of oral tolerance therapy with type II collagen suggest that nasal exposure may be even more effective in tolerance induction.62 However, the results of oral collagen in patient trials63 have not reflected the degree of success observed in animals and may reflect both the complexity of the pathology of RA and the difficulty in regulating, let alone modulating, the human diet. Combination therapy, where cytokine modulators are used to reinforce the induction of tolerance64, may prove to be a promising approach to human disease, particularly as the appropriate toleragen for RA still remains poorly defined.

TRANSGENIC MODELS The use of transgenic mice to examine the contributions of specific genes or proteins to the pathogenesis of arthritis represents a novel, powerful approach to the development of anti-arthritic therapies. Early in this technology, the appearance of arthritis in transgenic mice for no immediately obvious reason generated surprise, and fostered the hypothesis that genetic alterations expressed within the joint might lead to non-specific arthritis-like conditions. One such early model occurred in mice

52 P. H. Wooley

transgenic for the env-pX region of the human T cell leukaemia virus (HTLV) type 1 genome which developed a spontaneous chronic arthritis due to the expression of the tax gene.65 Ankle joints were most frequently affected, and pannus formation leading to severe erosions of cartilage and bone was observed after several months of disease. In affected joints, cytokine genes (IL-1b, IL-6, TNF-a TGF-b1, g-IFN and IL-2) were activated, leading to elevated serum cytokine levels, elevated IgG levels and production of rheumatoid factor and antibodies against type II collagen, Hsps, nuclear antigens and cardiolipin. Other abnormal immune responses include the increased expression of class II MHC antigens, and reactivity against type II collagen and Hsp. It is now thought that reactivity to connective tissue antigens may well be key in this model66, because collagen immunization can provoke arthritis onset or exacerbation, and the T cells infiltrating skin and joint lesions appear to be oligoclonal in nature and share a similar T cell receptor Vb profile with T cells isolated from joints of mice with CIA.66 However, this model has added to speculation that a retrovirus could be involved in the pathogenesis of RA, possibly by affecting synovial cell growth or regulation67 – 69 which could ultimately influence immune responses to cartilage antigens. Interestingly, the prevalence of HTLV-I Tax positivity among patients with RA has been reported to be three times higher than among healthy blood donors.70 Cytokine-directed immunotherapy for RA was considerably strengthened by the discovery that mice carrying 30 -modified hTNF-a transgenes exhibit deregulated TNFa expression and developed chronic inflammatory polyarthritis.71 At 2 weeks of age in TNF-a (Tg197) transgenic mice, moderate synovitis was present in rear paws and cartilage changes occurred in the absence of synovial infiltrate, with chondrocyte hyperproliferation, cartilage malformation and marked proteoglycan loss. Clinical disease was visible in rear paws from 3 weeks of age, with swelling of the ankle joints, while front paws became involved later. Synovitis, bony erosions and periosteal inflammation worsen, and eventually result in impaired mobility.72 TNF-a transgenics do not exhibit a broad autoimmune syndrome, but Tg197 mice have a markedly shortened lifespan and appear to die from cachexia. Because the arthritis develops as a result of the systemic overproduction of TNF-a, it was gratifying to find that anti-TNFa immunotherapy ameleriotes the joint disease and also extends the life of the Tg197 transgenic mice.71 Recent findings suggest that anti-TNFa in Tg197 mice may reverse existing structural damage, including synovitis and periosteal bone erosions, and this finding may correlate with the reduction in signs and symptoms of RA and the inhibition of the progression of structural joint damage seen in patients treated with inhibitors of TNFa.19,73,74 Because the roles of TNFa and IL-1b appear to be rather similar in inflammatory arthritis, the TNF transgenic model predicted that over-expression of IL-1 would also generate a model of joint disease. Transgenic mice that constitutively produce human IL-1a do indeed develop a chronic, inflammatory, symmetrical, erosive polyarthritis, with synovial hyperplasia, pannus formation and erosion of both cartilage and bone.75 An unusual feature of this murine model is the marked polymorphonuclear neutrophil (PMN) infiltration of the synovial joint. These cells expressed the Gr-1high phenotype, which has been associated with tissue damage and may account for the accelerated loss of cartilage in this model. The supporting observation that IL-1 receptor antagonist knockout mice develop arthritis and autoimmunity76 adds to the importance of this cytokine in the disease process. To date, preclinical studies involving IL-1 transgenic mice have not been reported, but cumulative evidence in other animal models has led to the development of IL-1 inhibitors that show efficacy in RA.77,78

Animal models and arthritis therapy 53

Transgenic mice have also proven useful in investigating the mechanisms of immunoregulation in RA. Transgenic mice expressing the DQA1 and DQB1 genes from an RA-predisposing (DQ8/DR4/Dw4) haplotype and the autologous murine class II genes ‘knocked out’ developed autoimmune responses and a severe polyarthritis following immunization with type II collagen.79 – 81 Control HLA-DQ8-H-2Ab0 mice developed neither disease nor anticollagen immunity. This finding implies that human class II HLA-DQ antigens may present type II collagen in an arthritogenic manner, and considerably strengthens the murine CIA model as representing a relevant feature of pathology of arthritis in man. HLA-DR4 (DRB1*0401) may also act to present either human or bovine type II collagen in an arthritogenic manner in transgenic mice.15 T cell and B cell autoimmune responses were seen against murine CII, and the DR4-restricted T cell response to human CII was focused on an immunodominant determinant within CII263-270 known to be key to collagen arthritis. Similar findings were observed in DR1 (DRB1*0101) transgenic mice immunized with human CII81, with the development of severe autoimmune and erosive arthritis accompanied by DR1-restricted immune responses to human CII. Recent findings show that DR1 and DR4 bind the CII(263-270) peptide in a nearly identical manner82, indicating that both HLA-DR4 and HLA-DR1 are capable of binding peptides derived from human CII, which could be significant to the pathology of RA. Myers et al83 have recently built on this observation to develop peptides capable of altering the immune response to collagen by disrupting the DR1restricted immune response. These peptides greatly reduced the incidence and severity of CIA in HLA-DR1 transgenic mice, and thus we may be positioned to see the development of an ‘RA vaccine’ in the near future. The obvious limitation to this approach is the (as yet unknown) prevalence of a DR1-restricted autoimmune response to collagen in the pathology of a general RA population. However, the feasibility of this vaccine therapy raises exciting prospects for the future. A surprising novel transgenic model has provoked many researchers to re-examine some of the basic concepts of autoimmunity in RA84, and might revolutionize our thinking on arthritis immunotherapy. The K/B £ N T cell receptor mouse model appeared when the KRN TCR transgenic mouse was crossed with the NOD (nonobese diabetes) strain.85 The progeny developed an aggressive spontaneous, symmetrical arthritis in peripheral joints with a pathology resembling human RA. Histological features included synovial infiltration, pannus formation, erosive disease and bone remodelling. The critical autoimmune reactivity has been shown to reside in the serendipitous specificity of the KRN Vb6 T cell receptor. Although initially characterized as reactive with a bovine ribonuclease peptide in context of the MHC antigen I-Ak, the KRN T cell receptor recognizes glucose-6-phosphate isomerase (G6PI) in the context of the NOD-derived I-Ag7 class II MHC molecule.86 This immune recognition gives rise to anti-G6PI autoantibodies, which appear critical to the development of joint disease, as passive transfer of anti-G6PI antibodies can result in the appearance of transient joint disease. The precise pathological relevance of an immune response to G6PI in the aetiology of arthritis is unclear87,88, because G6PI is ubiquitously expressed in all cells. The significance of this autoantibody in joint disease was further enhanced by the discovery of anti-G6PI autoantibodies in RA patients89, suggesting a common mechanism that may prove highly relevant to joint disease. However, recent findings suggest that no disease-specific pattern of antibody positivity to GPI exists in RA patients, and that the frequency of anti-G6PI immunity in RA is lower than previous estimates.90 – 92 It is too early to generalize upon the significance of the KB/N model as a useful tool in the study of the immunopathology of RA, and we may yet see therapeutic approaches based upon this discovery. But the recent doubts

54 P. H. Wooley

again emphasize the problems with interpreting animal models of human disease and raise the issue of whether transgenic manipulation (and the subsequent selective processes that permit a genetically altered animal to survive) can always represent true single-gene events. This concern will affect novel therapy development, as exemplified when the surprising discovery that disruption of the stromelysin-1 gene was without effect on susceptibility to collagen-arthritis in B10.RIII transgenic mice.93 This observation caused a marked loss of enthusiasm for this matrix metalloproteinase as a useful therapeutic target.

SUMMARY This chapter is not intended to represent a complete picture of the role of animal models in the development of anti-arthritic therapies, and there are several significant omissions beyond the scope of this current article. The list of publications alone that cite novel drugs and modalities from herbal medicines to stress exposure is beyond easy recollection. Hopefully, I have addressed several of the high points in drug discovery that have occurred during the last quarter century and seen the contribution of animal models to significant advances in the array of pharmaceutical agents now available to rheumatologists. Nevertheless, significant challenges remain: RA is still a disease of unknown aetiology, and none of the therapies described represents a cure. It will be interesting to see whether progress in animal models will eventually provide the profound insights required to conquer this condition. However, it appears highly likely that animal models will continue to generate novel targets for intervention in RA for the next 25 years.

REFERENCES *1. Trentham DE, Townes AS & Kang AH. Autoimmunity to type II collagen an experimental model of arthritis. Journal of Experimental Medicine 1977; 146: 857 –868. 2. Billingham MEJ. Adjuvant arthritis: the first model. In Henderson B, Edwards JCW & Pettipher ER (eds) Mechanisms and Models in Rheumatoid Arthritis. London: Academic Press, 1995, pp 389 –410. 3. Wooley PH, Seibold JR, Whalen JD & Chapdelaine JM. Pristane-induced arthritis. The immunologic and genetic features of an experimental murine model of autoimmune disease. Arthritis and Rheumatism 1989; 32: 1022–1030. 4. Kleinau S, Erlandsson H, Holmdahl R & Klareskog L. Adjuvant oils induce arthritis in the DA rat. I. Characterization of the disease and evidence for an immunological involvement. Journal of Autoimmunity 1991; 4: 871–880. 5. van Eden W, Hogervorst EJ, Hensen EJ et al. A cartilage-mimicking T-cell epitope on a 65K mycobacterial heat- shock protein: adjuvant arthritis as a model for human rheumatoid arthritis. Current Topics in Microbiology and Immunology 1989; 145: 27–43. 6. Brackertz D, Mitchell GF & Mackay IR. Antigen-induced arthritis in mice. I. Induction of arthritis in various strains of mice. Arthritis and Rheumatism 1977; 20: 841–850. 7. Howson P, Shepard N & Mitchell N. The antigen-induced arthritis model: the relevance of the method of induction to its use as a model of human disease. Journal of Rheumatology 1986; 13: 379 –390. 8. McCune WJ, Trentham DE & David JR. Gold does not alter the arthritic, humoral, or cellular responses in rats with type II collagen-induced arthritis. Arthritis and Rheumatism 1980; 23: 932–936. 9. Stuart JM, Myers LK, Townes AS & Kang AH. Effect of cyclophosphamide, hydrocortisone and levamisole on collagen-induced arthritis in rats. Arthritis and Rheumatism 1981; 24: 790 –794. 10. Wooley PH, Luthra HS, O’Duffy JD et al. Anti-type II collagen antibodies in rheumatoid arthritis. The influence of HLA phenotype. Tissue Antigens 1984; 23: 263 –269. 11. Clague RB, Firth SA, Holt PJ et al. Serum antibodies to type II collagen in rheumatoid arthritis: comparison of 6 immunological methods and clinical features. Annals of the Rheumatic Diseases 1983; 42: 537 –544.

Animal models and arthritis therapy 55 12. Bartholomew JS, Evanson JM & Woolley DE. Serum IgE anti-cartilage collagen antibodies in rheumatoid patients. Rheumatology International 1991; 11: 37 –40. 13. Iribe H, Kabashima H, Ishii Y & Koga T. Epitope specificity of antibody response against human type II collagen in the mouse susceptible to collagen-induced arthritis and patients with rheumatoid arthritis. Clinical and Experimental Immunology 1988; 73: 443–448. 14. Terato K, Shimozuru Y, Katayama K et al. Specificity of antibodies to type II collagen in rheumatoid arthritis. Arthritis and Rheumatism 1990; 33: 1493–1500. * 15. Ranges GE, Sriram S & Cooper SM. Prevention of type II collagen-induced arthritis by in vivo treatment with anti-L3T4. Journal of Experimental Medicine 1985; 162: 1105–1110. 16. Levitt NG, Fernandez-Madrid F & Wooley PH. Pristane induced arthritis in mice. IV. Immunotherapy with monoclonal antibodies directed against lymphocyte subsets. Journal of Rheumatology 1992; 19: 1342– 1347. 17. Banerjee S, Webber C & Poole AR. The induction of arthritis in mice by the cartilage proteoglycan aggrecan: roles of CD4 þ and CD8 þ T cells. Cellular Immunology 1992; 144: 347 –357. 18. van den Broek MF, van de Langerijt LG, van Bruggen MC et al. Treatment of rats with monoclonal antiCD4 induces long-term resistance to streptococcal cell wall-induced arthritis. European Journal of Immunology 1992; 22: 57–61. 19. Burmester GR, Horneff G & Emmrich F. Management of early inflammatory arthritis. Intervention with immunomodulatory agents: monoclonal antibody therapy. Baillie`re’s Clinical Rheumatol 1992; 6: 415–434. 20. Horneff G, Burmester GR, Emmrich F & Kalden JR. Treatment of rheumatoid arthritis with an anti-CD4 monoclonal antibody. Arthritis and Rheumatism 1991; 34: 129– 140. 21. Herzog C, Walker C, Muller W et al. Anti-CD4 antibody treatment of patients with rheumatoid arthritis: I. Effect on clinical course and circulating T cells. Journal of Autoimmunity 1989; 2: 627– 642. 22. Veale DJ, Reece RJ, Parsons W et al. Intra-articular primatised anti-CD4: efficacy in resistant rheumatoid knees. A study of combined arthroscopy, magnetic resonance imaging, and histology. Annals of the Rheumatic Diseases 1999; 58: 342 –349. 23. Taylor PC. Antibody therapy for rheumatoid arthritis. Current Opinion in Pharmacology 2003; 3: 323– 328. * 24. Haqqi TM, Anderson GD, Banerjee S & David CS. Restricted heterogeneity in T-cell antigen receptor V beta gene usage in the lymph nodes and arthritic joints of mice. Proceedings of the National Academy of Sciences of the USA 1992; 89: 1253–1255. 25. Haqqi TM, Qu XM, Anthony D et al. Immunization with T cell receptor V beta chain peptides deletes pathogenic T cells and prevents the induction of collagen-induced arthritis in mice. Journal of Clinical Investigation 1996; 97: 2849–2858. 26. Chiocchia G, Manoury B, Boissier MC & Fournier C. T cell-targeted immunotherapy in murine collageninduced arthritis. Clinical and Experimental Rheumatology 1993; 11(supplement 9): S15– S17. 27. Chiocchia G, Boissier MC & Fournier C. Therapy against murine collagen-induced arthritis with T cell receptor V beta-specific antibodies. European Journal of Immunology 1991; 21: 2899–2905. 28. Goldschmidt TJ, Jansson L & Holmdahl R. In vivo elimination of T cells expressing specific T-cell receptor V beta chains in mice susceptible to collagen-induced arthritis. Immunology 1990; 69: 508–514. 29. Matsuno H, Kadowaki KM, Suzuki H et al. T-cell receptor V beta 8 has no significant suppressive effect on collagen-induced arthritis. International Journal of Tissue Reactions 1993; 15: 1 –9. 30. Paliard X, West SG, Lafferty JA et al. Evidence for the effects of a superantigen in rheumatoid arthritis. Science 1991; 253: 325–329. 31. Cuesta IA, Sud S, Song Z et al. T cell receptor (V beta) bias in the response of rheumatoid arthritis synovial fluid T cells to connective tissue antigens. Scandinavian Journal of Rheumatology 1997; 26: 166–173. 32. Gudmundsson S, Ronnelid J, Karlsson-Parra A et al. T-cell receptor V-gene usage in synovial fluid and synovial tissue from RA patients. Scandinavian Journal of Immunology 1992; 36: 681 –688. 33. Wooley PH, Whalen JD, Chapman DL et al. The effect of an interleukin-1 receptor antagonist protein on type II collagen-induced arthritis and antigen-induced arthritis in mice. Arthritis and Rheumatism 1993; 36: 1305– 1314. 34. Geiger T, Towbin H, Cosenti-Vargas A et al. Neutralization of interleukin-1 beta activity in vivo with a monoclonal antibody alleviates collagen-induced arthritis in DBA/1 mice and prevents the associated acute-phase response. Clinical and Experimental Rheumatology 1993; 11: 515–522. 35. Piguet PF, Grau GE, Vesin C et al. Evolution of collagen arthritis in mice is arrested by treatment with antitumour necrosis factor (TNF) antibody or a recombinant soluble TNF receptor. Immunology 1992; 77: 510–514. * 36. Thorbecke GJ, Shah R, Leu CH et al. Involvement of endogenous tumor necrosis factor alpha and transforming growth factor beta during induction of collagen type II arthritis in mice. Proceedings of the National Academy of Sciences of the USA 1992; 89: 7375–7379.

56 P. H. Wooley * 37. Wooley PH, Dutcher J, Widmer MB & Gillis S. Influence of a recombinant human soluble tumor necrosis factor receptor FC fusion protein on type II collagen-induced arthritis in mice. Journal of Immunology 1993; 151: 6602–6607. 38. Graninger W & Smolen J. Treatment of rheumatoid arthritis by TNF-blocking agents. International Archives of Allergy and Immunology 2002; 127: 10 –14. 39. Maini RN. Heberden Oration, 1988. Exploring immune pathways in rheumatoid arthritis. British Journal of Rheumatology 1989; 28: 466–479. 40. Moreland LW, Bucy RP, Weinblatt ME et al. Immune function in patients with rheumatoid arthritis treated with etanercept. Clinical Immunology 2002; 103: 13–21. 41. Kuruvilla AP, Shah R, Hochwald GM et al. Protective effect of transforming growth factor on experimental autoimmune diseases in mice. Proceedings of the National Academy Sciences of the USA 1991; 88: 2918–2921. 42. Cooper WO, Fava RA, Gates CA et al. Acceleration of onset of collagen-induced arthritis by intraarticular injection of tumour necrosis factor or transforming growth factor-beta. Clinical and Experimental Immunology 1992; 89: 244–250. 43. Mihara M, Kotoh M, Nishimoto N et al. Humanized antibody to human interleukin-6 receptor inhibits the development of collagen arthritis in cynomolgus monkeys. Clinical Immunology 2001; 98: 319–326. 44. Choy EH, Isenberg DA, Garrood Tet al. Therapeutic benefit of blocking interleukin-6 activity with an antiinterleukin-6 receptor monoclonal antibody in rheumatoid arthritis: a randomized, double-blind, placebocontrolled, dose-escalation trial. Arthritis and Rheumatism 2002; 46: 3143–3150. 45. Yoshizaki K, Nishimoto N, Mihara M & Kishimoto T. Therapy of rheumatoid arthritis by blocking IL-6 signal transduction with a humanized anti-IL-6 receptor antibody. Springer Seminars in Immunopathology 1998; 20: 247–259. 46. Robbins PD, Evans CH & Chernajovsky Y. Gene therapy for rheumatoid arthritis. Springer Seminars in Immunopathology 1998; 20: 197– 209. 47. Le CH, Nicolson AG, Morales A & Sewell KL. Suppression of collagen-induced arthritis through adenovirus-mediated transfer of a modified tumor necrosis factor alpha receptor gene. Arthritis and Rheumatism 1997; 40: 1662–1669. 48. Quattrocchi E, Walmsley M, Browne K et al. Paradoxical effects of adenovirus-mediated blockade of TNF activity in murine collagen-induced arthritis. Journal of Immunology 1999; 163: 1000–1009. 49. Yin Z, Siegert S, Neure L et al. The elevated ratio of interferon gamma-/interleukin-4-positive T cells found in synovial fluid and synovial membrane of rheumatoid arthritis patients can be changed by interleukin-4 but not by interleukin-10 or transforming growth factor beta. Rheumatology (Oxford) 1999; 38: 1058–1067. 50. Lubberts E, Joosten LA, van Den BL et al. Adenoviral vector-mediated overexpression of IL-4 in the knee joint of mice with collagen-induced arthritis prevents cartilage destruction. Journal of Immunology 1999; 163: 4546–4556. 51. Cottard V, Mulleman D, Bouille P et al. Adeno-associated virus-mediated delivery of IL-4 prevents collagen-induced arthritis. Gene Therapy 2000; 7: 1930–1939. 52. Watanabe S, Imagawa T, Boivin GP et al. Adeno-associated virus mediates long-term gene transfer and delivery of chondroprotective IL-4 to murine synovium. Molecular Therapy 2000; 2: 147– 152. 53. Guery L, Chiocchia G, Batteux F et al. Collagen II-pulsed antigen-presenting cells genetically modified to secrete IL-4 down-regulate collagen-induced arthritis. Gene Therapy 2001; 8: 1855–1862. 54. Whalen JD, Lechman EL, Carlos CA et al. Adenoviral transfer of the viral IL-10 gene periarticularly to mouse paws suppresses development of collagen-induced arthritis in both injected and uninjected paws. Journal of Immunology 1999; 162: 3625–3632. 55. Banda NK, Vondracek A, Kraus D et al. Mechanisms of inhibition of collagen-induced arthritis by murine IL-18 binding protein. Journal of Immunology 2003; 170: 2100– 2105. 56. Saidenberg-Kermanac’h N, Bessis N, Deleuze V et al. Efficacy of interleukin-10 gene electrotransfer into skeletal muscle in mice with collagen-induced arthritis. The Journal of Gene Medicine 2003; 5: 164–171. 57. Yang G, Di C, Jiang M et al. Effect of interleukin I receptor antigonist gene therapy on arthritis induced by type II collagen in mice. Zhonghua Yi Xue Za Zhi 2002; 82: 990–994. 58. Robbins PD, Evans CH & Chernajovsky Y. Gene therapy for arthritis. Gene Therapy 2003; 10: 902–911. 59. Evans CH, Ghivizzani SC, Herndon JH et al. Clinical trials in the gene therapy of arthritis. Clinical Orthopaedics & Related Research 2000; 379: S300–S307. 60. Nakajima A, Seroogy CM, Sandora MR et al. Antigen-specific T cell-mediated gene therapy in collageninduced arthritis. Journal of Clinical Investigation 2001; 107: 1293– 1301. 61. Staines NA & Wooley PH. Collagen arthritis—what can it teach us? British Journal of Rheumatology 1994; 33: 798–807. 62. Higuchi K, Kweon MN, Fujihashi K et al. Comparison of nasal and oral tolerance for the prevention of collagen induced murine arthritis. Journal of Rheumatology 2000; 27: 1038–1044.

Animal models and arthritis therapy 57 63. Barnett ML, Kremer JM, St Clair EW et al. Treatment of rheumatoid arthritis with oral type II collagen. Results of a multicenter, double-blind, placebo-controlled trial. Arthritis and Rheumatism 1998; 41: 290–297. 64. Thorbecke GJ, Schwarcz R, Leu J et al. Modulation by cytokines of induction of oral tolerance to type II collagen. Arthritis and Rheumatism 1999; 42: 110–118. * 65. Habu K, Nakayama-Yamada J, Asano M et al. The human T cell leukemia virus type I-tax gene is responsible for the development of both inflammatory polyarthropathy resembling rheumatoid arthritis and noninflammatory ankylotic arthropathy in transgenic mice. Journal of Immunology 1999; 162: 2956–2963. 66. Kotani M, Tagawa Y & Iwakura Y. Involvement of autoimmunity against type II collagen in the development of arthritis in mice transgenic for the human T cell leukemia virus type I tax gene. European Journal of Immunology 1999; 29: 54–64. 67. Nakajima T, Aono H, Hasunuma T et al. Overgrowth of human synovial cells driven by the human T cell leukemia virus type I tax gene. Journal of Clinical Investigation 1993; 92: 186–193. 68. Kawakami A, Nakashima T, Sakai H et al. Inhibition of caspase cascade by HTLV-I tax through induction of NF-kappaB nuclear translocation. Blood 1999; 94: 3847–3854. 69. Mori N, Shirakawa F, Abe M et al. Human T-cell leukemia virus type I tax transactivates the interleukin-6 gene in human rheumatoid synovial cells. Journal of Rheumatology 1995; 22: 2049–2054. 70. Zucker-Franklin D, Pancake BA & Brown WH. Prevalence of HTLV-I Tax in a subset of patients with rheumatoid arthritis. Clinical and Experimental Rheumatology 2002; 20: 161–169. * 71. Keffer J, Probert L, Cazlaris H et al. Transgenic mice expressing human tumour necrosis factor: a predictive genetic model of arthritis. EMBO Journal 1991; 10: 4025–4031. 72. Shealy DJ, Wooley PH, Emmell E et al. Anti-TNF-alpha antibody allows healing of joint damage in polyarthritic transgenic mice. Arthritis Research 2002; 4: R7. 73. Lipsky PE, van der Heijde DM, St Clair EW et al. Infliximab and methotrexate in the treatment of rheumatoid arthritis. Anti-tumor necrosis factor trial in rheumatoid arthritis with concomitant therapy study group. New England Journal of Medicine 2000; 343: 1594–1602. 74. Moreland LW, Schiff MH, Baumgartner SW et al. Etanercept therapy in rheumatoid arthritis. A randomized, controlled trial. Annals of Internal Medicine 1999; 130: 478 –486. 75. Niki Y, Yamada H, Seki S et al. Macrophage- and neutrophil-dominant arthritis in human IL-1 alpha transgenic mice. Journal of Clinical Investigation 2001; 107: 1127–1135. 76. Iwakura Y. Roles of IL-1 in the development of rheumatoid arthritis: consideration from mouse models. Cytokine Growth Factor Reviews 2002; 13: 341 –355. 77. Arend WP. The mode of action of cytokine inhibitors. Journal of Rheumatology 2002; 65(Supplement): 16–21. 78. Bresnihan B. The safety and efficacy of interleukin-1 receptor antagonist in the treatment of rheumatoid arthritis. Seminars in Arthritis and Rheumatism 2001; 30(5 supplement 2): 17– 20. * 79. Nabozny GH, Baisch JM, Cheng S et al. HLA-DQ8 transgenic mice are highly susceptible to collageninduced arthritis: a novel model for human polyarthritis. Journal of Experimental Medicine 1996; 183: 27 –37. * 80. Zanelli E, Krco CJ, Baisch JM et al. Immune response of HLA-DQ8 transgenic mice to peptides from the third hypervariable region of HLA-DRB1 correlates with predisposition to rheumatoid arthritis. Proceedings of the National Academy Sciences of the USA 1996; 93: 1814–1819. 81. Gabay C & Arend WP. Treatment of rheumatoid arthritis with IL-1 inhibitors. Springer Seminars in Immunopathology 1998; 20: 229 –246. 82. Rosloniec EF, Whittington KB, Zaller DM & Kang AH. HLA-DR1 (DRB1*0101) and DR4 (DRB1*0401) use the same anchor residues for binding an immunodominant peptide derived from human type II collagen. Journal of Immunology 2002; 168: 253–259. 83. Myers LK, Sakurai Y, Tang B et al. Peptide-induced suppression of collagen-induced arthritis in HLA-DR1 transgenic mice. Arthritis and Rheumatism 2002; 46: 3369–3377. 84. Hirano T. Revival of the autoantibody model in rheumatoid arthritis. Nature Immunology 2002; 3(4): 342–344. 85. Kouskoff V, Korganow AS, Duchatelle V et al. Organ-specific disease provoked by systemic autoimmunity. Cell 1996; 87: 811–822. * 86. Matsumoto I, Staub A, Benoist C & Mathis D. Arthritis provoked by linked T and B cell recognition of a glycolytic enzyme. Science 1999; 286: 1732– 1735. 87. Matsumoto I, Maccioni M, Lee DM et al. How antibodies to a ubiquitous cytoplasmic enzyme may provoke joint-specific autoimmune disease. Nature Immunology 2002; 3: 360–365. 88. Kyburz D, Carson DA & Corr M. The role of CD40 ligand and tumor necrosis factor alpha signaling in the transgenic K/BxN mouse model of rheumatoid arthritis. Arthritis and Rheumatism 2000; 43: 2571–2577.

58 P. H. Wooley 89. Schaller M, Burton DR & Ditzel HJ. Autoantibodies to GPI in rheumatoid arthritis: linkage between an animal model and human disease. Nature Immunology 2001; 2: 746 –753. 90. Matsumoto I, Lee DM, Goldbach-Mansky R et al. Low prevalence of antibodies to glucose-6-phosphate isomerase in patients with rheumatoid arthritis and a spectrum of other chronic autoimmune disorders. Arthritis and Rheumatism 2003; 48: 944–954. 91. Herve CA, Wait R & Venables PJ. Glucose-6-phosphate isomerase is not a specific autoantigen in rheumatoid arthritis. Rheumatology (Oxford) 2003; 42: 986–988. 92. Kassahn D, Kolb C, Solomon S et al. Few human autoimmune sera detect GPI. Nature Immunology 2002; 3: 411–412. 93. Mudgett JS, Hutchinson NI, Chartrain NA et al. Susceptibility of stromelysin 1-deficient mice to collageninduced arthritis and cartilage destruction. Arthritis and Rheumatism 1998; 41: 110–121.