throbber
http://www-ermm.cbcu.cam.ac.uk
`
`expert reviews
`
`on molecular medicine
`
`w
`.J
`Structure-function analysis and the molecular
`
`
`UJ
`C:
`
`
`
`
`origins of anti-DNA antibodies in systemic lupus
`erythematosus
`fl>
`(1)
`
`·-
`
`·-"'C
`0
`
`
`
`
`Jatinderpal Kalsi, Chelliah T. Ravirajan, Anisur Rahman and
`.c
`David A. Isenberg
`
`
`
`
`
`Patients with the autoimmune rheumatic disease systemic lupus erythematosus
`�
`
`
`
`(SLE or 'lupus') develop a wide variety of clinical and serological manifestations
`
`
`
`
`
`including the presence of antibodies to double-stranded DNA (dsDNA), which
`<C
`
`
`
`
`
`
`are often diagnostic and potentially pathogenic. In this review, we have examined Z
`
`
`
`
`the links between the structure and function of anti-dsDNA antibodies, 't
`
`
`
`
`
`their clinical associations. We have also reviewed studies involving ;:;
`emphasising
`
`
`
`
`
`animal models, the analysis of human antibody sequences and studies of, and C:
`
`
`
`using, computer modelling and crystal structure.
`<C
`
`....
`
`·-
`
`Systemic lupus erythematosus (SLE or 'lupus') is
`
`
`
`characterised by thickening of the skin), patients
`
`
`
`
`a major autoimmune rheumatic disease that is
`
`
`with SLE do not develop antibodies to the
`
`
`
`characterised clinically by photosensitive rashes
`DNA-binding enzyme Sci 70 (also known as
`
`and arthritis; in most cases the kidneys, lungs and
`
`
`topoisomerase 1). Furthermore, the relatively
`
`
`
`central nervous system are affected by the disease.
`
`
`
`
`restricted autoantibody profile that is typical of
`
`
`
`
`Its clinical diversity is, apparently, matched by its
`
`
`patients with SLE leads to the conclusion that
`
`
`serological diversity (see Table l; tabOOldil).
`
`random polyclonal activation of B cells cannot be
`
`
`
`Antibodies [immunoglobulins (Igs) produced by
`
`
`
`solely responsible for the production of anti­
`
`
`
`B lymphocytes (B cells)] that are found in patients
`
`
`DNA antibodies (Ref. 2). Precisely which of
`
`
`with SLE appear to target a range of 'self antigens'
`
`
`
`these SLE-associated autoantibodies are likely to
`
`
`
`(host-derived antigens). However, the diversity
`
`
`
`be pathogenic remains to be determined, and
`
`
`of the antibodies found in patients with SLE
`
`
`represents a considerable challenge.
`
`is actually rather narrow, given that a
`
`Anti-dsDNA antibodies
`
`
`
`typical mammalian cell contains physiologically
`
`
`significant quantities of as many as 2000 different
`
`
`Among the autoantibodies that are present in the
`
`
`proteins, which are all potential self antigens
`
`serum of patients with SLE, those that bind to
`
`
`(Ref. 1). For example, unlike patients with
`
`
`dsDNA remain of paramount interest. These
`
`
`scleroderma (an autoimmune disease that is
`
`
`antibodies were first identified in the serum of
`
`David A. Isenberg (Corresponding author)
`
`
`ARC Diamond Jubilee Professor of Rheumatology at University College London, Centre for
`
`
`
`
`
`
`
`
`
`
`Rheumatology /Bloomsbury Rheumatology Unit, Department of Medicine, University College
`
`
`
`
`
`London, Arthur Stanley House, 50 Tottenham Street, London WlP 9PG, UK, Tel: +44 (0)171 380
`
`
`9230; Fax: +44 (0)171 380 9278; E-mail: d.isenberg@ucl.ac.uk;
`
`
`
`
`
`Departmental website: http://www.ucl.ac.uk/ medicine /bloomsbury-rheumatology /
`
`Jatinderpal Kalsi, Chelliah T. Ravirajan and Anisur Rahman
`
`
`
`
`Centre for Rheumatology /Bloomsbury Rheumatology Unit, Department of Medicine, University
`
`
`
`
`
`
`
`
`
`
`College London, Arthur Stanley House, 50 Tottenham Street, London WlP 9PG, UK
`
`1
`{99)00042-3a.pdf (short code: txt001 dil); 16 February 1999
`
`Accession information:
`ISSN 1462-3994 ©1999 Cambridge University Press
`Pfizer v. Genentech
`IPR2017-01488
`
`Genentech Exhibit 2023
`
`

`

`http://www-ermm.cbcu.cam.ac.uk
`
`expert reviews
`in molecular medicine
`
`-— LIJ
`
`Table 1. Summary of the frequency of serum antibodies (and the antigens they are
`
`detected with) that are commonly detected in patients with systemic lupus
`
`
`emthematosus (SLE) (tab001dil)
`
`
`Frequency ofpatients studied who have detectable antibodies (“/o)
`
`
`Published studies
`Series at the Bloomsbury
`Antigens the lgs bind to
`
`Rheumatology Unitb
`
`(worldwide) (Ab type)a
`
`
` 34 (for lgG);12 (for lgM)
`20—50
` Cardiolipin
`Not studied
`C1q
`20—45
`
`55
`dsDNA
`40—90
`21
`
`
`Fc IgG (RF)
`~25
`Not studied
`Histone
`30—80
`hsp 70
`5—40
`
`
`hsp 90
`5—50
`5 (for IgG); 35 (for lgM)
`La
`10—15
`13
`
`
`14
`LAC
`10—20
`97
`‘Nucleus’ (ANA)
`>90
`
`
`20
`RNP
`20—35
`32
`Ro
`30—40
`
`
`10
`Sm
`5—30
`Not studied
`ssDNA
`s70
`
`
`20
`Thyroid
`S35
`
`
`
`a For further details about the antigens used, see Table 13 in Ref. 106.
`
`
`b The Bloomsbury Rheumatology Unit is at University College, London, UK.
`
`Abbreviations used: Ab = antibody; ANA = anti-nuclear antibody; C1q = a complement protein; dsDNA =
`
`double—stranded DNA; hsp 70 and hsp 90 = two types of human heat shock protein; histone = the protein
`associated with DNA; lg = immunoglobulin (antibody); LAC = lupus anti-coagulant; RF = rheumatoid factors,
`antibodies (usually lgM) that bind to lg and are produced by patients with rheumatoid arthritis but also with
`
`
`other diseases; ssDNA= single—stranded DNA; Sm, RNP, R0 and La = ribonuclear proteins; thyroid = an
`
`
`extract of human thyroid.
`
`
`nSL
`
`
`
`Anti-DNAantibodies
`
`
`
`5 2
`
`patients with SLE over 40 years ago in four
`different laboratories (reviewed in Ref. 3). The
`likely involvement of these antibodies in the
`pathogenesis of human SLE, and in animal models
`of SLE, is indicated by (1) the close links between
`disease ’activity’ and levels of anti—dsDNA
`antibodies in the serum of many, though not all,
`patients; (2) the elution (removal and collection)
`of these antibodies from the kidneys of patients
`with SLE and lupus-prone mice; (3) direct
`evidence of these antibodies being associated with
`pathogenicity in isolated rat perfusion systems
`in which kidneys are dissected from the rat and
`their function is maintained artificially for a
`few hours) and mice with severe combined
`immunodeficiency (SCID); and (4) the fact that
`although antibodies to single—stranded DNA
`(ssDNA) are frequently found in the relatives of
`patients with SLE, those that bind to dsDNA are
`virtually never detected.
`
`Clinical studies
`
`Many papers published during the past 20
`years (reviewed by Spronk and colleagues in
`
`Ref. 4) have concluded that levels of anti-
`
`dsDNA antibodies [usually measured using
`enzyme-linked immunosorbent assays (ELISAs)
`or radioimmunoassays (RIAs)] generally reflect
`Clinical disease activity. This observation appears
`to be particularly true of renal (kidney) disease,
`and most of the evidence that anti—dsDNA
`
`antibodies are pathogenic has been collected from
`studies of the kidney. Thus, high levels of anti-
`dsDNA antibodies, as measured using the Farr
`assay, which detects high-avidity antibodies
`(those that bind strongly to their target antigens),
`and low values of the complement enzyme
`marker CHSO were found predominantly in
`patients with lupus nephritis (i.e. who have
`kidney inflammation associated with SLE; Ref. 5).
`In contrast, antibodies to ssDNA are not specific
`for patients with SLE, being present, for example,
`in many individuals with infectious diseases.
`Lloyd and Schur (Ref. 6) found that complement
`depletion and raised titres of anti-dsDNA
`antibodies were associated more closely with
`renal than non-renal exacerbations in SLE. Ter
`
`Borg and colleagues (Ref. 7), in a prospective
`
`Accession information: (99)OOO42-3a.pdf (Short code: txtOO1dil); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`
`

`

`http://www-ermm.cbcu.oam.ac.uk
`
`expert reviews
`in molecular medicine
`
`study of 72 patients, showed that active lupus
`nephritis was usually associated with high
`titres of
`anti—dsDNA antibodies. More
`
`recently, Okamura and colleagues (Ref. 8) have
`demonstrated a close relationship between renal
`disease activity (assessed by biopsy) and the
`isotype of Ig (e.g. IgG compared with IgM)
`produced that bound DNA: activity correlated
`with IgG reactive against dsDNAbut not with IgG
`reactive against ssDNA or with IgM reactive
`against either dsDNA or ssDNA. Bootsma and
`colleagues (Ref. 9), using the concept of a rise in
`levels of anti-dsDNA antibodies as a means of
`
`predicting a clinical relapse, showed that treating
`such patients with high levels of prednisolone (30
`mg / day) reduced the relapse rate, compared with
`a control group, who were treated with either
`lower doses of prednisolone or no steroids.
`
`Assessment of disease activity in SLE
`A major problem with many of these earlier
`studies was the unsatisfactory nature of the
`indices of disease activity that were used to assess
`SLE. Validated and reliable global-score indices
`(i.e. score systems in which activity in each system
`is ’lumped together’) have been developed
`only in the past decade. However, with a
`disease as diverse as SLE, an activity index that
`demonstrates ’at a glance’ the degree of disease
`activity in each of the major organs or systems
`has obvious advantages. The British Isles Lupus
`Assessment Group (BILAG) has described and
`validated such a system, based on the ’physician’s
`intention to treat’ principle (Ref. 10). Thus, eight
`organs or systems are distinguished, allowing the
`easy correlation of activity in a particular organ
`or system with any serological marker. Using this
`system in a serial, longitudinal study of 14 Afro-
`Caribbean patients with SLE, antibodies to
`dsDNA were shown to correlate with renal
`
`disease, cardiopulmonary disease and global
`scores, but not with musculoskeletal, central
`nervous system or haematological involvement.
`Blood samples taken over periods of follow—up
`in the range of 3—15 years were analysed in this
`study (Ref. 11).
`
`Historical evidence that anti-dsDNA
`
`antibodies can be pathogenic in SLE
`Detection alone of anti-DNA antibodies in the
`
`lesinSLE
`
`Anti-DNAantibod
`
`at ’the scene of the crime’. Koffler and colleagues
`(Ref. 12) eluted IgG, complement and IgM from
`the kidneys of patients who had died from lupus
`nephritis. These eluates possessed anti-nuclear-
`binding activity; although a specific test for anti-
`DNA binding was not performed, the anti-
`nuclear-binding activity of the eluted antibodies
`could be partially inhibited by the addition of
`dsDNA, suggesting that at least some anti-dsDNA
`antibodies were present.
`In a study (Ref. 13) of over 40 families affected
`by SLE (where 21% of 147 relatives had antibodies
`to ssDNA), only two relatives had antibodies
`to dsDNA, strongly implicating anti-dsDNA
`antibodies, but not the anti-ssDNA antibodies, in
`the disease process (Ref. 14).
`
`Spontaneous disease models of SLE;
`pathogenicity of anti-DNA antibodies
`Some strains of mice spontaneously develop
`autoimmune disorders that have many of the
`features that are typical of human SLE (Ref. 15).
`The most commonly studied strains of mice with
`murine lupus are: (1) New Zealand black (NZB),
`(2) BWF1 [NZB x New Zealand white (NZW) F1],
`(3) MRL/++, (4) MRL-lpr/lpr, (5) MRL/lpr, (6)
`BXSB, and (7) SNFl [(NZB) x (SWR) F1] (for
`a comparison of their features see Table 2;
`tabOOZdil). These mice produce elevated levels of
`total Igs and autoantibodies (such as anti-dsDNA)
`and are thought to develop nephritis and arteritis
`as a result of deposition of immune complexes in
`the kidneys or arteries. Like patients with SLE,
`these mice present a diversity of disease patterns.
`In NZB and BWF1 mice, disease occurs primarily
`in the females. The disease in NZB mice is
`
`characterised mostly by a type of haemolytic
`anaemia that is Coombs positive (with anti-red-
`cell antibodies produced), and this can be
`associated with variable production of anti-
`nuclear antibodies (ANAs). The disease in
`BWF1 mice is characterised by the production
`of ANAs and immune-complex-mediated
`glomerulonephritis (in their kidneys). In MRL/
`lpr mice, the lupus disease more equally affects
`male and female mice, whereas in BXSB mice, the
`disease affects males, because of the influence of
`
`a Y—linked gene (Ref. 15).
`
`kidneys of patients with SLE does not prove such
`MRL/ ++ and MRL/ lpr mice are used as models
`antibodies were responsible for the development
`of this complication; however, it does place them
`of SLE, and genetically differ at the lpr locus,
`—3
`Accession information: (99)OOO42—3a.pdf (short code: txtOO‘idii); 16 February 1999
`ISSN 1462—3994 ©1999 Cambridge University Press
`
`The role of the lpr gene in mouse models
`of SLE
`
`

`

`
`
`Anti-DNAantibodiesinSLE
`
`http://www—ermm.cbcu.cam.ac.uk
`
`expert reviews
`in molecular medicine
`
`§ T
`
`able 2. Features of disease in lupus-prone mouse models (tab002dil)
`
`Mouse strain;
`MHC haplotype
`
`New Zealand black
`(NZB); H-2d
`
`Major clinical features
`
`Haemolytic anaemia,
`glomerulonephritis,
`lymphomas.
`
`Survival time for 50%
`of animals in typical
`group; sex of animal
`most affected
`
`18 months; both sexes
`affected equally.
`
`BWF1 [F1 of (NZB
`x New Zealand white
`(NZW)]§ H-2d’Z
`
`MRL—lpr/Ipr, H-2k
`
`Severe immune-complex-
`mediated nephritis.
`
`7—8 months; females.
`
`Lymphoproliferation,
`immune-complex-mediated
`nephritis, rheumatoid
`arthritis, vasculitis.
`
`2—4 months; females.
`
`Major immunological
`features
`
`Production of anti-erythrocyte
`antibodies, hyperproduction of
`lgM, generalised lymphocyte
`dysfunction.
`
`Production of anti-nuclear and
`anti-DNA antibodies,
`generalised lymphocyte
`dysfunction.
`
`Production of anti-nuclear
`antibodies and rheumatoid
`factors, proliferation of Ly1+
`cells“, generalised lymphocyte
`dysfunction.
`
`MRL‘“; H—2k
`
`As for MRL-lpr/lpr but less
`severe.
`
`18 months; females.
`
`As for M RL-lpr/Ipr but less
`severe.
`
`
`
`BXSB; H-2h
`
`Haemolytic anaemia,
`lymphadenopathy,
`glomerulonephritis.
`
`2—4 months; males.
`
`Moth-eaten; H-2b
`
`Hair loss, glomerulonephritis,
`increased susceptibility
`to infections.
`
`1 month; both sexes
`affected equally.
`
`Palmerston—North;
`H—2q
`
`Polyarteritis nodosa,
`immune-complex-mediated
`nephritis.
`
`Swan; H-2k
`
`Mild glomerulonephritis.
`
`11 months; females.
`
`Production of anti-DNA
`antibodies, anti-NTA and
`anti-erythrocyte antibodies,
`thymic atrophy occurs earlier
`than normal.
`
`Production of anti-DNA
`antibodies, anti-NTA and anti-
`erythrocyte antibodies,
`immunosuppression.
`
`Production of anti-DNA
`antibodies, hyperactivity of B
`lymphocytes.
`
`18 months; both sexes
`affected equally.
`
`Production of anti-DNA
`antibodies, thymic atrophy
`occurs earlier than normal.
`
`SNF1; H-ZL"d
`
`Severe glomerulonephritis.
`
`4—8 months; females.
`
`Production of anti-DNA and
`anti-nucleosome antibodies.
`
`a Ly1+ cells = mouse T lymphocytes (T cells) that have Ly1 antigens on their cell surface, a T-helper subset.
`Abbreviations used: H-2 = mouse major histocompatibility complex (MHC) class I; lgM = isotype of
`immunoglobulin (lg); NTA = natural thymocytotoxic antibody.
`
`which is mutated in lpr mice and is an autosomal
`recessive gene (Ref. 16). In the homozygous state,
`lpr causes an immunological
`illness that
`is characterised by lymphoproliferation
`and accelerated autoimmunity. MRL/ ++ mice,
`which have two copies the wild-type (unmutated)
`gene at the lpr locus, develop an autoimmune
`
`syndrome with a much longer survival time of
`the mice. Congeneic mice on various genetic
`backgrounds that have two copies of the mutated
`lpr gene (such as C57BL/ 6-lpr/lpr, Bé-lpr/Zpr,
`C3I-I/HeI-lpr/lpr and AKR-lpr/ lpr) all develop
`’spontaneous’ lymphoproliferation and produce
`autoantibodies. Despite the fact that anti-DNA
`
`—A
`
`ccession information: (99)OOO42-3a.pdf (short code: txtOO1dil); 16 February 1999
`ISSN 1462—3994 ©1999 Cambridge University Press
`
`

`

`http://www—ermm.cbcu.cam.ac.uk
`
`expert reviews
`in molecular medicine
`
`— a
`
`ntibodies are produced by, and found in, all of
`the strains of mice, only MRL / lpr mice (with one
`wild-type and one mutant lpr gene) develop
`severe immune-complex renal disease, which is
`responsible for the death of these mice by 6—9
`months of age. Therefore, the underlying or
`primary mechanism of autoimmunity in the MRL
`model lies in the MRL/ ++ background, and the
`lpr gene appears to act by accelerating and
`increasing the severity of the disease.
`The lpr gene is now known to encode a
`defective configuration of the Fas receptor
`(Ref. 17), the normal form of which mediates a
`signalling pathway that initiates apoptosis
`(programmed cell death; Ref. 18). Fas is a cell-
`surface protein that plays a major role in the
`induction of apoptosis in lymphoid cells. Mice
`with mutations in lpr and / or lprcg, the gene(s)
`encoding Fas in mice (Ref. 19), are characterised
`by prolonged survival of B cells and other features
`that are similar to those found in patients with
`SLE, suggesting that in these animals interference
`with apoptosis might be important in the
`pathogenesis of their autoimmune disease (see
`below). Because of the involvement of lpr in
`apoptosis, which is important to T—cell function,
`we recently investigated whether MRL / lpr mice
`had increased activity in the T—helper (Th) subset
`of T cells (Ref. 20). T cells in the lymph nodes (of
`both mice and humans) produce soluble factors
`that enhance B—cell activation and, in turn,
`
`Control of anti-DNA antibodies in mice
`
`cause the accumulation of large numbers of
`lymphocytes in the spleen and lymph nodes.
`These T cells are either conventional CD4+ T cells
`or CD4‘ CD8‘ T cells (also known as ‘double
`negative’ T cells). In MRL/ lpr mice, a failure of
`apoptosis of CD4+ T cells in the periphery of the
`thymus allows self-reactive T cells to persist and
`by T cells and cytokines
`ultimately to drive autoantibody production
`T cells probably play an important role in these
`by B cells (Ref. 21). Our studies revealed no
`mouse models of SLE because experimental
`significant difference in the extent of apoptosis in
`elimination of Th T cells in SNF1 mice virtually
`the organs of MRL/lpr mice compared with
`prevented disease, and the removal of Th cells in
`normal (non-autoimmune) BALB/ c mice at one
`BWF1 mice also improved disease outcome (Ref.
`month of age. At this age, the MRL/ lpr mice had
`25). CD4+ T cells, which can induce B cells to
`low levels of serum anti—ssDNA antibodies and
`secrete cationic IgG anti-dsDNA antibodies, have
`anti-dsDNA antibodies, and showed no detectable
`been cloned from SNF1 mice, which develop
`impairment in renal functions. This observation
`an SLE—like autoimmunity (Ref. 26). Studies on
`confirms that in MRL/ lpr mice, at one month,
`the role of
`those T cells
`that express
`when the distribution of apoptotic cells was
`CD40 ligand (CD4OL) in the development of
`normal, there was no detectable lupus disease.
`glomerulonephritis in SNF1 mice have shown that
`Initially, in the MRL / lpr mice, there were no global
`blocking the interaction between CD4OL on the
`defects in the negative selection of the T—cell
`pathogenic Th cells and CD40 on the lupus B cells,
`repertoire, and the positive selection of subsets
`— 5
`Accession information: (99)OOO42-3a.pdf (short code: txtOOi dil); 16 February 1999
`ISSN 1462—3994 ©1999 Cambridge University Press
`
`
`
`Anti-DNAantibodiesinSLE
`
`of CD4+ T cells and CD8+ T cells appeared to
`be normal (Ref. 22). Failure of apoptosis of
`self-reactive CD8+ T cells can then lead to
`
`downregulation of CD8 and persistence as
`CD4“ CD8‘ T cells, which contribute to the
`lymphadenopathy. It should be noted that in the
`MRL/ lpr mice, the size of the thymus increased
`in parallel with the augmented activity of the Th
`T cells (Ref. 23).
`
`Anti-DNA antibodies in mice
`
`In mice, the genetic basis of the ability to produce
`anti—dsDNA antibodies has not been completely
`defined. In most strains, nephritis is associated
`with the co—inheritance of several genes including
`major histocompatibility complex (MI 1C) genes.
`In the SNF1 mouse, nephritis is linked to the I—Acl
`locus of the normal SWR parent, whereas in BWF1
`mice, it is linked to the I-Eb chain from the NZW
`parent, a different MHC gene.
`Each of these autoimmune mouse strains
`
`has a polyclonal B-cell hyperactivity (many
`clones of B cells are affected) that causes
`hypergammaglobulinaemia (high levels of IgG
`antibodies) and an increased number of antibody-
`forming cells, including those that produce
`antibodies that bind to nuclear antigens. Although
`the B cells that are capable of producing the high-
`affinity anti-dsDNA antibodies are present in
`normal mice, they do not proliferate and actually
`produce such antibodies. Experimentally,
`to investigate the relationship between
`autoimmunity and Fas-mediated apoptosis (see
`below), using appropriate fusion—partner cells
`(Ref. 24), autoantibody—producing cells can be
`’immortalised’ to produce Igs in culture.
`
`

`

`http://www—ermm.cbcu.cam.ac.uk
`#m
`cardiolipin and to nucleic acids (Fig. 1; fig001dil; —l
`at a particular window of time, delayed the
`Ref. 32). Together, these data suggest that DNA w
`expansion of autoimmune memory B cells, and
`itself might not be the immunogen that is
`resulted in long—term therapeutic benefits in the
`responsible for inducing what are commonly :
`mice (Ref. 27). Thus, it is theoretically possible that
`known as anti-DNA antibodies.
`I—
`patients with SLE might benefit from treatment
`with monoclonal antibodies that are reactive with
`CD4 or CD40.
`
`expert reviews
`In molecular medicine
`
`tibodies
`
`Non-mammalian DNA as an eliciting
`agent
`As reviewed elsewhere (Ref. 33), mammalian
`DNA is poorly immunogenic in mammals. One
`of the key questions, therefore, concerns the
`identity of the agent(s) responsible for eliciting
`the anti—DNA-antibody response in patients with C
`SLE. Several candidates have been suggested, and ‘5
`have been reviewed below.
`Experiments have shown that bacterial DNA <
`is able to induce strong antibody responses in 2
`mice. Gilkeson and colleagues (Refs 34, 35) have 0
`proposed that bacterial DNA could be the
`I
`immunogen that induces the production of "I':
`antibodies that are reactive to DNA in patients :
`with SLE. When bacterial DNA was injected into <
`healthy mice, antibodies that lacked specificity for
`mammalian DNA were mostly found; these
`antibodies lacked the molecular characteristics
`
`Studies of mouse models of SLE have suggested
`a role for two cytokines, interleukin 6 (IL-6) and
`interleukin 10 (IL-10), in the pathophysiology of
`SLE. Treatment of BWF1 mice with an anti-IL-10
`monoclonal antibody delayed the onset of
`autoimmune manifestations and the production
`of autoantibodies (Ref. 28). Similarly, treatment
`of BWFl mice with an anti-IL—6-receptor antibody
`lowered the production of IgG autoantibodies and
`suppressed the development of autoimmune
`disease (Ref. 29). Again, these data suggest that
`the use of appropriate monoclonal antibodies in
`patients with SLE might be beneficial (Ref. 30).
`
`In vivo stimulation of anti-DNA antibody
`production
`The anti-DNA antibody response in SLE has been
`shown to be heterogeneous (Ref. 31). Serum-
`derived antibodies and monoclonal anti—DNA
`antibodies recognised epitopes that occur in
`different nucleic acids (such as phosphodiester
`groups separated by three adjacent carbon atoms;
`see Fig. 1; fig001dil) on apparently diverse
`molecules such as phospholipids, proteins,
`polysaccharides and cell—membrane structures
`(reviewed in Ref. 31). These observations were not
`due either to non—specific binding of low-affinity
`antibodies or merely to a charge interaction,
`because it has
`also been shown that
`anti—’DNA’ antibodies do not necessarily bind
`polynucleotides that have similar patterns of ionic
`charge (Ref. 31). Furthermore, IgG antibodies that
`have high affinity for DNA (Ref. 31) can show the
`same degree of polyreactivity (i.e. ability to react
`with different epitopes) as some low-affinity IgM
`anti—DNA antibodies.
`
`(notably arginine residues in the VH CDR3
`region) that are typical of the anti-dsDNA
`antibodies that are found in patients with SLE
`(Ref. 35). Autoantibodies in the serum of
`patients with SLE can bind to a widely shared
`bacterial epitope (on DNA), though these
`antibodies were mainly directed against ssDNA
`rather than dsDNA.
`
`a human dsDNA
`Studies using BK,
`polyoma virus, have produced data that are more
`promising. Hyperimmunisation of BALB/ c mice
`(Ref. 36) with this virus has been shown to induce
`the production of anti-DNA antibodies, and
`analysis of the variable-region genes of the
`antibodies revealed that the induced antibodies
`resembled the anti—DNA antibodies that are
`
`This polyreactivity of DNA antibodies could
`be due to multiple binding sites in variable regions
`in individual Igs or to the same (or similar)
`epitopes in different antigens. Furthermore,
`hybridoma cells (a fusion of antibody-producing
`cells and a transformed cell line) that were
`prepared from mice that had been immunised
`with conjugates of cardiolipin (a phospholipid)
`and protein have been shown to produce
`monoclonal antibodies that bound both to
`—6
`Accession information: (99)OOO42-3a.pdf (short code: txtOO1diI); 16 February 1999
`ISSN 1462-3994 ©1999 Cambridge University Press
`
`characteristic of lupus in mice. Rekvig and
`colleagues (Ref. 37) have shown that the injection
`of BK into young lupus—prone mice (NZB x NZW
`F1)before the natural onset of the disease induced
`a strong and persistent anti-dsDNA response,
`which is more typical of the response found in
`patients with SLE. In a subsequent report
`(Ref. 38), the same group showed that in vivo
`expression of a single DNA-binding protein,
`the polyoma virus T antigen, was sufficient to
`initiate the production of anti-dsDNA and anti-
`histone antibodies (histones are the protein core
`
`

`

`http://www—ermm.cbcu.cam.ac.uk
`
`expert reviews
`in molecular medicine
`
`Phosphodiester
`group
`
`Three adjacent
`carbon atoms
`
`Putative
`shared epitope
`for anti-DN'A-I
`antibody binding
`
`
`
`Anti-DNAantibodiesinSLE
`
`'
`o—é—R
`HC’
`I
`o
`H20\ O_5_R
`
`H
`
`|R
`
`
`
`BASE
`
`20
`
`\ ,
`c
`
`CH
`'
`HC’
`|
`HC|)\C/
`0 H2
`
`Single strand of DNA
`
`Cardiolipin (phospholipid)
`
`Potential shared epitope between DNA and cardiolipin
`Expert Reviews in Molecular Medicine @1999 Cambridge University Press
`
`Figure 1. Potential shared epitope between DNA and cardiolipin.Anti-DNA antibodies found in the serum
`of patients with systemic lupus erythematosus (SLE) have been shown to bind to many antigenic targets
`including single- and double-stranded DNA (ssDNA and dsDNA) and phospholipids such as cardiolipin. In
`addition, mouse monoclonal antibodies from mice immunised with cardiolipin can bind to nucleic acids and
`cardiolipin (Ref. 32). This cross-reactivity might be due to a shared epitope in their primary sequence, consisting
`of one or more phosphodiester groups (Ref. 107) separated by three adjacent carbon atoms (shown here for
`ssDNA and cardiolipin). Abbreviation used: R = side chain. Modified from Ref. 107 (figOO1dil).
`
`responsible for the production of anti-DNA
`antibodies.
`
`around which dsDNA ’wraps’). They also
`provided further evidence that DNA alone is
`not immunogenic in Vivo, thus adding support
`to the increasingly popular theory that DNA Nucleic-acid—protein conjugates as
`that is complexed to histones in the form of
`eliciting agents
`nucleosomes is the key immunogenic particle Nucleosomes are the fundamental repeating units
`— 7
`Accession information: (99)OOO42-3a.pdf (short code: tXtOO‘l dil); 16 February 1999
`ISSN 1462—3994 ©1999 Cambridge University Press
`
`

`

`non-autoimmune mice by immunisation with
`complexes formed from mammalian DNA and the
`Fus-1 protein from the parasite Trypanosoma cruzi
`(Ref. 50). However, the significance of these
`observations to the aetiology of human SLE
`remains uncertain.
`
`expert reviews
`in molecular medicine
`
`(O
`
`http://www- ermm. cbcu.cam. ac uk
`—m
`several mammalian histone peptides and also
`of chromatin; they are generated during apoptosis
`other components that are common target
`by internucleosomal cleavage of the chromatin
`antigens in autoimmune diseases, such as
`(Ref. 39) Bell and colleagues (Ref. 40) have
`Sm-D antigens, ubiquitinated HZA antigen and
`demonstrated that B-cell hybridoma cells from
`poly(ADP——ribose)antigens (Ref. 48) Recently, we
`patients with SLE in culture released nucleosomes
`have shown that, in the mouse strain MRL/ lpr,
`that stimulated proliferation and Ig synthesis in
`normal B cells of mice.
`which is genetically susceptible to lupus, the
`In mouse models of SLE, the onset of the
`injection of as little as 1 ug of mouse histone—RNA
`complex induced increased antibody responses to
`autoimmune response has been shown to be
`murine histones, dsDNA and ubiquitinated H2A,
`characterised by the early emergence of antibodies
`and exaggerated the disease in vivo in young mice
`that recognise conformational epitopes of the
`(Ref. 49).
`nucleosome particles (Refs 41, 42). As the immune
`Antibodies to dsDNA were also induced in
`response progressed, autoantibodies that reacted
`to dsDNA or histones became apparent. In
`addition, in mice with lupus, nucleosomes were
`a major immunogen for inducing Th T cells that
`could stimulate pathogenic autoantibodies
`(Ref. 43).
`Our research group has produced from one
`patient with SLE a panel of five IgG (and five IgM)
`human hybridoma-derived monoclonal anti-
`DNA antibodies; two of these IgG antibodies
`bound to both dsDNA and histones, whereas none
`of the IgM antibodies did (Ref. 44). Moreover, the
`binding of these human monoclonal antibodies
`to histones was enhanced by the presence of DNA,
`which was also detectable in the supernatants of
`the hybridomas. In a subsequent study, we
`analysed the pathogenic properties of monoclonal
`human hybridoma anti-DNA IgG antibodies by
`implanting and growing the hybridoma-secreting
`cells in the peritoneal cavity of SCID mice. Only
`the IgG antibodies that bound to both dsDNA and
`histones (or nucleosomes) deposited in the renal
`glomeruli of the mice, producing renal damage
`(Refs 45, 46). These data imply that some anti-
`dsDNA antibodies have the capacity to bind
`directly to the renal tissue and cause damage,
`perhaps without the help of complement.
`Conjugates of nucleic acid and proteins, where
`the nucleic acids can be DNA or other nucleic
`acids, and the proteins can be histone or other
`proteins, can also induce anti-DNA antibodies. For
`example, in normal, non-SLE-susceptible mice,
`bacterial DNA that is complexed to methylated
`bovine serum albumin (BSA) in a suitable
`immunogenic. A recent experimental model of
`adjuvant can induce the production of high titres
`lupus (Ref. 54) that used peptide immunisation
`of IgG anti—DNA antibodies (Ref. 47). However,
`of normal rabbits extols the concept of epitope
`anti—DNA antibodies induced1n this way do not
`spreading as one of the mechanisms that might
`bind mammalian dsDNA, despite their reactivity
`generate anti-DNA antibodies. Our own work,
`with mammalian histone proteins that are
`however, does not substantiate this model
`complexed with mammalian RNA. These IgG
`antibodies react with mammalian histones,
`(Ref. 55).
`# 8
`Accession information: (99)OOO42-3a.pdf (short code: txtOO1diI); 16 February 1999
`ISSN 1462—3994 ©1999 Cambridge University Press
`
`Molecular mimicry and cross-reactivity
`A microorganism that possesses both foreign
`epitopes and epitopes that mimic autoantigens
`has the potential to elicit an autoimmune response
`in a human or animal that is exposed to it. Indeed,
`Diamond and colleagues (Refs 51, 52) have shown
`that autoantibodies were generated as part of the
`immune response to a bacterial immunogen.
`This group described 99D.7E, a cross—reactive
`monoclonal mouse IgG antibody that bound to
`both dsDNA and phosphorylcholine, which is a
`component of the bacterial cell wall. Although the
`antibody 99D.7E provided partial protection
`against the microorganism, it was pathogenic to
`the kidney. It is also possible that immune
`responses might arise against a particular
`component because of its close association with
`another component. Rabbits that were immunised
`with purified rabbit RNA polymerase 1 (Ref. 53)
`produced antibodies that were reactive against the
`RNA polymerase 1 enzyme and also rabbit nucleic
`acids. It is possible that these antibodies against
`nucleic acids were induced not by cross-reactive
`epitopes on RNA polymerase 1, but because the
`physical association between RNA polymerase 1
`and nucleic acids rendered the nucleic acids
`
`EA
`
`
`
`nti-DNAantibodiesi
`
`

`

`http://www—ermm.cbc:u.cam.ac.uk
`#m
`coding for Fas or the Fas ligands. However, levels
`Other stimuli of anti-dsDNA-antibody
`_I
`of soluble Fas are elevated in a minority of patients
`production
`who have SLE only (Ref. 57).
`Abnormalities of the immune system in human
`Bcl—2 is one of a family of proto-oncogene
`SLE no doubt serve to enhance or perpetuate
`products; the principal Bcl-2 protein is unusual
`the anti—DNA-antibody response. It has been
`among such products in that it enhances the
`suggested that several other factors might also
`survival of lymphoid cells by interfering
`’assist’ in the generation of the autoantibody
`with apoptosis, rather than by promoting cell
`response.
`proliferation. Much fundamental research work
`is in progress to explore how apoptosis is
`regulated; some members of the Bcl-2 f

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket