You are on page 1of 14

Immunomodulators

Arthur Kavanaugh and David H Broide


94

METHOTREXATE
CONTENTS SUMMARY OF IMPORTANT CONCEPTS

>> At present, immunomodulator therapy (other than traditional


• Introduction 1643 allergen protein immunotherapy and anti-IgE) in the field of
• Methotrexate 1643 asthma and allergy remains investigational
• Immunophilin-binding agents and calcineurin inhibitors: >> Double-blind studies of IVIG in asthma have not demonstrated
cyclosporin, tacrolimus, and pimecrolimus 1645 a therapeutic benefit
• Intravenous immunoglobulin 1648 >> Anti-TNF, methotrexate, and cyclosporin have shown a mod-
est benefit in some early-phase studies in asthma
• DNA-based therapies 1649
>> Anti-IL-4 and anti-IL-5 therapies have not proved effective in
• Mycobacterial vaccines 1653 the treatment of persistent asthma
• Cytokine-based therapy 1654 >> Immunotherapy with cytosine phosphorothioate guanosine
• Conclusions 1655 DNA conjugated to a protein allergen has shown promise in
animal models of asthma and in small-scale human studies,
but further large-scale studies are needed to determine its
safety and effectiveness

■ INTRODUCTION ■
Recognition of the importance of the Th2 lymphocyte as an orches-
trator of the immune and inflammatory response associated with
■ METHOTREXATE ■
asthma has focused attention on the use of immunomodulator therapy. PHARMACOLOGY, STRUCTURE,
Immunomodulators that have been successfully used in other immuno-
AND METABOLISM
logically mediated diseases (e.g., methotrexate in rheumatoid arthritis;
cyclosporin in transplant rejection; intravenous immunoglobulin (IVIG) Methotrexate can be administered orally or parenterally.1 Oral administra-
in idiopathic thrombocytopenic purpura) are not currently routinely tion results in a bioavailability of approximately 85%, although this may
used in the therapy of asthma or allergy, either because of the lack of decrease at higher doses.1 Subcutaneous or intramuscular dosing results in
studies showing a consistent beneficial effect and/or the potential side equivalent, near complete bioavailability. About half of absorbed methotrex-
effects related to therapy. Second-generation DNA-based immunomod- ate is protein bound, and may be displaced by agents such as non-steroidal
ulators have shown significant promise in animal models of asthma, but anti-inflammatory drugs (NSAIDs), sulfonamides, and others. The elimi-
whether this potential will be borne out in human studies is at present nation half-life of methotrexate is approximately 8 hours, and excretion
unknown. In this chapter we review the safety and efficacy of currently occurs predominantly via glomerular filtration. Indeed, renal function is a
available (methotrexate, cyclosporin, IVIG) and investigational (DNA- critical factor in methotrexate pharmacokinetics, and hence its toxicity.1
based, anti-cytokine) immunomodulators. Traditional allergen protein Methotrexate may be considered as both drug and prodrug, as it is
immunotherapy (Chapter 95) and novel immunomodulation strategies rapidly taken up and metabolized to a series of polyglutamated deriva-
targeting adhesion molecules (Chapter 9) and chemokines (Chapter 11) tives that persist intracellularly for longer periods of time.1 Methotrexate
are covered in other chapters. polyglutamated intracellular metabolites interfere with the same metabolic

1643
F THERAPEUTICS

N N NH2
CH3 Table 94.1 Adverse effects potentially associated with
methotrexate
N N
H
Common
NH2
HOOC N
Oral ulcerations
COOH O ‘Post-dose’ reactions (arthralgia, fatigue)
Methotrexate Nausea, diarrhea

N N NH2 Less common

Abnormal liver function tests


H N N Skin rash
OH
HOOC N H Uncommon
METHOTREXATE

COOH O Pulmonary toxicity


Folic acid Myelosuppression
Alopecia
Fig. 94.1. Chemical structures of methotrexate and folic acid.
Increased risk of infection
Hepatic fibrosis
pathways as does the parent compound.1 Serum methotrexate concentra-
Lymphoma (Epstein–Barr virus associated)
tions do not correlate with clinical efficacy in non-malignant conditions,
whereas levels of intracellular polyglutamated methotrexate do.1
Methotrexate and folic acid are structurally similar (Fig. 94.1). Meth-
otrexate and its polyglutamated metabolites avidly bind the active site of by amino-imidazole-carboxy-amido-ribonucleotide (AICAR) trans-
the enzyme dihydrofolate reductase, thereby blocking the conversion of formylase, leading to the increased release of adenosine; and (2) in-
dietary folic acid to its reduced form, tetrahydrofolate. Depletion of the terference with transmethylation reactions, such as the methylation
reduced form of folate leads to reduced synthesis of thymidine, thereby of homocysteine to methionine.1 The precise mechanisms by which
impairing DNA synthesis. Methotrexate also inhibits other enzymes AICAR inhibition results in increased extracellular concentrations of
relevant to nucleotide synthesis, including thymidylate synthetase. adenosine remain to be defined, but through interactions with specific
Allelic variation in the genes encoding enzymes affecting methotrexate’s receptors (A1, A2a, A2b, A3) on various cell types, adenosine exerts
uptake and metabolism, and also in various potential targets affecting potent and diverse antiinflammatory actions.
their sensitivity to methotrexate, has been described. There is substantial
interest in the potential for pharmacogenetic profiling as a means to
ADVERSE EVENTS
enhance methotrexate’s efficacy while minimizing its toxicity.
Methotrexate can be associated with a number of adverse effects
(Table 94.1). Because dosing regimens, indications, and relevant factors
IMMUNOLOGIC EFFECTS
such as renal function vary significantly among published reports, it is
Inhibition of cell replication may be the key therapeutic mechanism of difficult to assign exact figures to the expected prevalence of these adverse
methotrexate in neoplastic conditions. Lymphocytes divide rapidly in the effects. Many side effects, including myelosuppression, are dose depend-
course of the immune response, and thus immunosuppression may be one ent and occur infrequently at the doses typically used in non-malignant
mechanism of action of methotrexate in various immunologic diseases. diseases. Some side effects, such as pulmonary reactions, are idiosyncratic.
However, several observations have suggested that other mechanisms may Many side effects are more commonly seen early in the treatment course,
be relevant in non-malignant conditions. For example, in contrast to its use or with changes in dose regimen. Many side effects can be attenuated
in the treatment of malignancies, the lower doses of methotrexate com- by decreasing methotrexate dosages or by the supplemental use of folic
monly used in rheumatoid arthritis (RA) are infrequently associated with acid.2 Although the administration of folic or folinic acid, particularly in
myelosuppression or clinical signs of systemic immunosuppression. In ad- large doses, can reduce efficacy, there appears to be consensus that the use
dition, supplementation with low doses of folic acid may obviate some of of folic acid in low doses (e.g., approximately 1 mg/day) is a cost-effective
the toxicity seen in RA patients receiving methotrexate without signifi- way to reduce adverse effects in patients taking methotrexate while at the
cantly attenuating its clinical efficacy.1,2 In vivo studies have shown that same time not substantially reducing efficacy. Moreover, supplemental
methotrexate can induce various immunomodulatory effects, including folic acid reverses the increase in serum homocysteine that is observed
effects on cellular immunity, humoral immunity, and inflammation.1,3,4 with methotrexate therapy – a potentially important consideration for
Two relevant mechanisms underlie the anti-inflammatory effect of patients with other risk factors for the development of atherosclerotic
methotrexate at the doses typically used in non-malignant conditions: disease. The potential hepatic toxicity of methotrexate may vary with
(1) inhibition of the intermediate step in purine metabolism catalyzed factors such as frequency of dosing (weekly versus daily), concomitant

1644
Immunomodulators 94
alcohol use, and differential host susceptibility. Significant hepatic fibrosis
Table 94.2 Recommendations for monitoring for
is rarely seen in patients who do not drink alcohol. Screening for other
methotrexate hepatic safety in patients with rheumatoid
factors that might predispose to liver injury, such as hepatitis, is now rou-
arthritis (RA).
tine. The American College of Rheumatology have developed guidelines
for monitoring liver toxicity (Table 94.2).5 Guidelines call for the evalu-
ation of selected liver function tests at regular intervals during therapy. If A Baseline
liver function tests do become elevated, this often reverses upon decreas- 1. Tests for all patients
a. Liver blood tests (aspartate aminotransferase [AST],
ing the dose of methotrexate. Putative markers of active hepatic fibrosis,
alanine aminotransferase [ALT], alkaline phosphatase,
for example serum concentrations of the aminoterminal propeptide of
albumin, bilirubin, serologic tests for
type III collagen, are under investigation.6
hepatitis B and C
Potential pulmonary toxicities related to methotrexate use have been
b. Other tests (serum creatinine, complete blood count)
somewhat difficult to define clearly. The largest literature addressing this 2. Pre-treatment liver biopsy only for patients with:
issue comes from the use of methotrexate in patients with RA, 0.5–5% a. Prior excessive alcohol consumption
of whom are reported to develop pulmonary toxicity.7 Much of the dif- b. Persistently abnormal baseline AST
ficulty defining the condition or establishing the true incidence arises c. Chronic hepatitis B or C infection
from the lack of pathognomonic clinical, laboratory, or radiological fea-
B Monitor AST, ALT, albumin at 4–8-week intervals
tures of ‘methotrexate pneumonitis.’ Common clinical characteristics

IMMUNOPHILIN-BINDING AGENTS AND CALCINEURIN INHIBITORS


may include shortness of breath, cough, and fever. Interstitial or alveolar C Perform liver biopsy if:
infiltrates on chest radiograph, and hypercellularity with lymphocytic 1. Five of nine determinations of AST within a given 12-month
predominance on bronchoalveolar lavage, are considered consistent interval (6 of 12 if performed monthly) are abnormal
with the diagnosis. Differentiation from pulmonary complications of 2. Albumin decreases below normal
the underlying disease may be suggested by the acute or subacute on- D If results of liver biopsy are:
set of symptoms in a previously stable patient beginning methotrexate 1. Roenigk grade I, II, or IIIa, resume methotrexate and
relatively recently; half of affected patients started methotrexate in the monitor
previous 8 months. However, differentiation from an infectious process 2. Roenigk grade IIIB or IV, discontinue methotrexate
is difficult. Corticosteroids may shorten the duration or improve the E. Discontinue methotrexate in patients with persistent liver test
symptoms of methotrexate pneumonitis in some patients. abnormalities who refuse liver biopsy
Because it is a teratogen, methotrexate is strictly contraindicated in
(Adapted from Kremer J, Alarcon G, Weinblatt M, et al. Clinical, laboratory,
pregnancy (US Food and Drug Administration category X). radiographic and histopathologic features of methotrexate-associated lung
injury in patients with rheumatoid arthritis. Arthritis Rheum 1997; 40:1829,7
with permission. Copyright Wiley-Liss Inc, a subsidiary of John Wiley &
CLINICAL STUDIES IN ASTHMA AND ALLERGY
Sons, Inc.).
In the late 1980s, open-label observations suggested that methotrexate
may have a corticosteroid-sparing effect in corticosteroid-dependent
adults with asthma.8 This was subsequently assessed in controlled trials 15 to 25 mg. Concurrent use of folic acid (1 mg/day) may obviate some
in adults and children9–11 and in systematic reviews12,13 which concluded adverse effects. Regular monitoring of selected liver function tests, blood
that there is evidence to support the ability of methotrexate to act as a cell counts, and renal function is useful to prevent adverse effects.
corticosteroid-sparing agent in corticosteroid-dependent asthma; how-
ever, the magnitude of the dose reduction was relatively modest (mean ■ IMMUNOPHILIN-BINDING AGENTS
reduction of 4.1–4.4 mg in the daily oral dose of prednisone equivalent).
Although this may represent a reduction in total oral corticosteroid dose AND CALCINEURIN INHIBITORS:
by 20% or more, the clinical significance of this effect can be debated. CYCLOSPORIN, TACROLIMUS,
Methotrexate, typically given in a dose of 15 mg/week, was generally
well tolerated. Longer courses of therapy (e.g. 6 months or more) might
AND PIMECROLIMUS ■
have been associated with a greater benefit.12
PHARMACOLOGY AND METABOLISM
CURRENT RECOMMENDATIONS FOR USE Cyclosporin A (CsA) is an 11 amino acid cyclic peptide initially isolated
in the 1970s from a soil organism (Tolypocladium inflatum) (Fig. 94.2).
IN CLINICAL PRACTICE
It was found to have substantial immunosuppressive properties, par-
Although it is widely used as an immunomodulatory therapy in autoim- ticularly inhibition of helper T-cell function.14 CsA blocks T cells via
mune conditions, the use of methotrexate as a therapeutic agent for pa- inhibition of calcineurin, which subsequently downregulates cytokines
tients with allergic disease may be most appropriate in patients with severe such as IL-2 that are critical for T-cell activation. Tacrolimus (previously
asthma that has been refractory to or dependent on oral corticosteroids. referred to as FK506; Fig. 94.2), also initially isolated from a soil organ-
In ‘corticosteroid-resistant’ asthma, methotrexate has been shown to have ism (Streptomyces tsukubanesis) in 1984, and pimecrolimus are structur-
a modest corticosteroid-sparing effect. The greatest guidance for the use ally distinct from CsA, but share its mechanisms of action.14–17 CsA
of methotrexate in non-malignant conditions comes from its use in RA. and tacrolimus have been introduced for clinical use in the setting of
Typical weekly doses for patients with normal renal function range from transplantation and for autoimmune diseases. Topical formulations of

1645
F THERAPEUTICS

O O TCR
H
N N N O
N N
O OH O IP3
H
O N N
H H
O O
Calcium
N N C
N NH O I
O O Calcineurin Calcineurin
I
Cyclosporin A T

O P NF-AT NF-AT P
Dephosphorylation
IMMUNOPHILIN-BINDING AGENTS AND CALCINEURIN INHIBITORS

O O
O Cytokine
H OH
transcription
O
N

H Fig. 94.3. Mechanism of action of cyclosporin (cyclosporin A) and


tacrolimus. Antigen stimulation of the T-cell receptor (TCR) leads to the
O O generation of inositol triphosphate (IP3), increases in cytosolic calcium, and
OH
formation of an activated calcineurin. Calcineurin is a phosphatase which
O dephosphorylates the phosphorylated transcription factor for nuclear factor of
O
activated T cells (NF-AT), allowing NF-AT to translocate to the nucleus to
induce expression of cytokine genes. Cyclosporin (C) and tacrolimus (T) bind
to their respective immunophilins (I) to block the phosphatase action of cal-
O
cineurin and thus prevent the expression of cytokine genes induced by NF-AT.
Tacrolimus

Fig. 94.2. Chemical structures of cyclosporin (cyclosporin A) and Steady-state whole blood levels of 400 ng/mL are often associated with
tacrolimus. trough concentrations of 300 ng/mL – a reasonable therapeutic target.
In non-transplant conditions the doses are typically lower. These agents
are lipophilic, and are highly bound to lipoproteins in the circulation.
tacrolimus and pimecrolimus have been approved for use in patients Topical tacrolimus and pimecrolimus have minimal systemic absorp-
with moderate to severe atopic dermatitis. Another structurally similar tion, with low serum levels being detectable in approximately 20–25%
compound, sirolimus (previously referred to as rapamycin), has a distinct of treated patients.17
mechanism of action and has not yet been introduced for clinical use.15 CsA and tacrolimus are eliminated primarily via biotransformation in
CsA and tacrolimus have variable pharmacokinetics and narrow the cytochrome P450 system (CYP3A) in the gut wall and liver. Impair-
therapeutic windows, making close monitoring mandatory.16 Monitor- ment of hepatic function reduces elimination of metabolites, whereas re-
ing of trough whole blood concentrations is standard for the use of these nal failure has an insignificant effect. A number of other medications are
drugs in solid organ transplantation. Monitoring of drug concentrations also metabolized by this same pathway, and therefore drug–drug interac-
is not standard in autoimmune conditions such as rheumatoid arthritis tions may significantly affect concentrations of CsA and tacrolimus.
and psoriasis, partly because of the lower doses used and because of the
lack of correlation of drug concentrations with relevant clinical outcomes
IMMUNOLOGIC EFFECTS
in non-transplant diseases.16 The microemulsion formulation of CsA has
more consistent bioavailability than with older preparations. Tacrolimus CsA, tacrolimus, and pimecrolimus exert their more prominent immu-
has about 100 times more immunosuppressive activity in vitro than CsA; nomodulatory effects on T-cell function. After being taken up by the cell,
therapeutic whole blood trough concentrations of tacrolimus are about these agents bind to cytosolic proteins called immunophilins: CsA binds
20 times lower than those for CsA. to cyclophilin, and tacrolimus to FK-binding protein (FKBP). The com-
CsA and tacrolimus are both available as intravenous and oral prepa- bination of drug and immunophilin binds to and inhibits calcineurin,
rations. Typical intravenous doses are about one-third of the oral dose. a calcium–calmodulin-dependent serine/threonine phosphatase that
In transplantation, doses of CsA are typically in the range of 5–8 mg/ activates various transcriptional regulatory factors (Fig. 94.3).15,18 A
kg/day, whereas for tacrolimus a typical dose would be 0.3 mg/kg/day. In key target of calcineurin is nuclear factor of activated T cells (NF-AT).
transplantation, dosing is guided by whole blood drug concentrations. Upon activation, NF-AT activates the transcription of genes encoding

1646
Immunomodulators 94
cytokines critical for T-cell function, particularly IL-2. Other T-cell tremor, and dysesthesias, have been reported in up to 20% of transplant
cytokines blocked by these agents include IL-3, IL-4, IL-5, GM-CSF, patients. Hypertrichosis, with hair growth typically on the face, arms, and
and IFN-γ. Because helper T cells play a central role in the initiation and shoulders, has been reported in up to 50% of transplant patients, as has
propagation of immune reactions, inhibition of helper T-cell function gingival hyperplasia. These effects are observed less commonly among
also exerts secondary modulatory effects on various other components non-transplant patients receiving lower doses. Glucose intolerance,
of the immune and inflammatory responses. In addition, CsA and tac- which is often compounded by the concurrent use of corticosteroids,
rolimus exert direct immunomodulatory effects on other cell types, for may relate to a direct effect of these drugs on pancreatic islet cell func-
example blocking degranulation and inhibition of the transcriptional tion, and seems to be more of a problem with tacrolimus than with CsA.
activation of genes encoding IL-3 and IL-5 by mast cells and basophils.18 Cholestasis and increases in hepatic transaminase concentrations have
Other changes that have been observed with immunophilin-binding also been observed.
agents include downregulation of the expression of the high-affinity As with any agents that suppress the immune response, potential
receptor for IgE. side effects related to the interference with normal immunosurveillance,
namely increased susceptibility to infection and malignancy, are a con-
cern with CsA and tacrolimus. In the setting of allograft transplanta-
ADVERSE EVENTS
tion, where the situation is made more difficult because of the concur-
Potential adverse effects occurring with oral and parenteral CsA and rent use of other immunosuppressants, cases of opportunistic infections
tacrolimus are similar (Table 94.3). One of the most common and sig- have been observed. In addition, cases of lymphoproliferative disease

IMMUNOPHILIN-BINDING AGENTS AND CALCINEURIN INHIBITORS


nificant adverse effects is nephrotoxicity. Whereas nephrotoxicity is seen and other malignancies have been reported among transplant patients.
similarly with both agents, hypertension, which can be a contributing In non-transplant recipients, such as patients with autoimmune diseases,
factor in renal dysfunction, may occur less frequently with tacrolimus.19 there does not appear to be an increased incidence of infection or malig-
Both an acute, reversible nephrotoxicity, related to alterations in intrare- nancy related to the use of these agents.20
nal hemodynamics, as well as a chronic irreversible renal dysfunction can Topical tacrolimus and pimecrolimus are generally well tolerated, with
be seen. The arteriolar vasoconstriction relevant to the acute decrease in the most common side effects being local burning upon application.17
renal function seen with these agents may be mitigated by calcium chan- In 2006, the US Food and Drug Administration mandated a ‘black box’
nel blocking agents. Hypertension can be an insidious problem, even at warning on the product label for topical tacrolimus and pimecrolimus
lower doses, and may limit long-term use of these drugs. concerning a potential risk for treatment-related malignancy. Analysis
A variety of adverse effects may occur with CsA and tacrolimus that, of clinical trial and pharmacovigilance data, however, would not seem
although not life-threatening, can be bothersome enough to preclude to support an increased risk of malignancy related to the use of these
long-term use of these drugs. Neurologic symptoms, including headache, agents.17

Table 94.3 Adverse effects potentially associated with CLINICAL STUDIES IN ASTHMA AND ALLERGY
cyclosporin and tacrolimus
To date, the calcineurin inhibitors have been studied mainly in two allergic
conditions: asthma and atopic dermatitis. In a blinded, 12-week crossover
Common study, treatment with CsA, at an initial dose of 5mg/kg/day, resulted in an
improvement in FEV1 of 12% compared to placebo.21 No attempt at cor-
Renal insufficiency (acute and chronic)
ticosteroid sparing was investigated, but fewer increases in corticosteroid
Hypertension dose were needed among the CsA-treated patients.21 In a follow-up study,
Hypertrichosis 39 patients with corticosteroid-dependent asthma experienced a 25% re-
Headache duction in corticosteroid dose while on CsA.22 The potential utility of CsA
Gingival hypertrophy
in asthma would appear to be supported by studies showing that it can
attenuate the allergen-induced late-phase reaction.23 Inhibition of the late-
Hyperuricemia phase asthmatic response is associated with attenuation of allergen-induced
Less common increases in IL-5, GM-CSF, eotaxin, and eosinophilia in the airway.
Not all studies of CsA have had a positive outcome. In one study24
Paresthesias neither improvements in lung function nor any corticosteroid-sparing
Tremor
effect could be demonstrated; hence CsA remains an investigational
treatment in asthma. Novel routes of administration, such as nebuliza-
Nausea tion, are under investigation.
Glucose intolerance T cells appear to have an important role in the initiation and mainte-
nance of atopic dermatitis (AD), providing a rationale for the use of CsA
Uncommon and tacrolimus in this condition. In one double-blind, placebo-controlled,
8-week crossover study CsA significantly improved the extent and
Increased risk of infection
severity of AD.25 Of note, despite the short duration of the study, at the
Increased risk of lymphoproliferative diseases dose of CsA used (5 mg/kg/day), 20 of the 33 patients developed adverse
Cholestasis effects, compared to 8 of 33 receiving placebo. Other studies have sug-
gested that CsA may have a beneficial effect in AD, but concerns about

1647
F THERAPEUTICS

toxicity remain. Because much of the relevant immune reaction driving enhanced clearance of endogenous IgG, anti-idiotype suppression, and
AD takes place locally in the dermis, topical calcineurin inhibitor admin- neutralization of super antigens.30 Which, if any, of these effects may
istration could theoretically minimize adverse effects while maintaining be useful in the treatment of asthma or allergy is currently not known.
its therapeutic effect. A topical preparation of CsA was not effective,26 In vitro studies have shown that IVIG inhibits cytokine-dependent
presumably related to poor absorption. In contrast, tacrolimus and pime- lymphocyte proliferation, as well as cytokine (IL-2, IL-4) production by
crolimus have been successfully developed as local immunomodulatory T lymphocytes.
agents for the treatment of AD.17,27 Trials with both agents have shown
notable improvements in signs and symptoms, with minimal toxicity
Immunoglobulin E
(see Chapter 62).
Inhibition of IgE production by B cells is one postulated mechanism
CURRENT RECOMMENDATIONS FOR USE by which IVIG may exert an immunomodulating effect in asthma.31,32
This effect of IVIG is postulated to occur through co-ligation of the
IN CLINICAL PRACTICE
B cell Fcγ RIIB receptor and the B-cell antigen receptor, with resultant
Recommendations for the use of calcineurin inhibitors in clinical practice negative signaling in B cells.32 IVIG could thus provide an off signal to
come from evidence accumulated to date in allergic diseases and extrapo- the B cell to inhibit B-cell proliferation and immunoglobulin produc-
lation from other diseases. CsA remains an investigational agent in the tion. In vitro, IVIG inhibits IgE production by B cells.31 In open-label
INTRAVENOUS IMMUNOGLOBULIN

treatment of asthma, in which its use is based on the wider experience studies IVIG has been shown to reduce the immediate skin reactivity to
with this agent in autoimmune diseases such as RA and psoriasis. In those allergen.33 Although there is some experimental evidence to suggest that
conditions, therapy with CsA is often started with a dose of the micro- IVIG may reduce IgE levels in vivo,33 a reduction in IgE is not noted in
emulsion formulation of about 2.5 mg/kg/day, given in divided doses. the majority of studies which have investigated this immunomodulatory
Doses are typically titrated upwards in increments of 0.5 mg/kg/day at property of IVIG.34,35
about 8-week intervals. Long-term doses higher than 4 mg/kg/day are
not typically used outside the transplant setting. The utility of monitoring
Sinopulmonary infections
trough concentrations of CsA in non-transplant patients remains unde-
fined. Longitudinal monitoring of renal function and blood pressure is IVIG might theoretically improve asthma by reducing the incidence of
required for patients using CsA or tacrolimus chronically. If the baseline sinopulmonary infections. Evidence for this has not adequately been ad-
creatinine increases by 30% or more, the dose should be reduced.28 Simi- dressed in current studies with IVIG.
larly, significant increases in blood pressure necessitate CsA dose reduc-
tion or the introduction of antihypertensive agents (i.e. calcium channel
ADVERSE EVENTS
blocking agents such as felodipine, amlodipine), with attention given to
potential drug interactions. Patients should be monitored for signs and IVIG therapy is generally safe and well tolerated. Side effects such as nau-
symptoms of infections, neurologic side effects, and other adverse effects. sea, headache, backache, chills, and flushing occur in approximately 5–10%
The use of topical tacrolimus and pimecrolimus for atopic dermatitis of IVIG infusions (Table 94.4)36 and usually resolve with interruption of
has increased, and the clinical indications are reviewed in Chapter 62. At the infusion and resumption at a slower rate. More serious adverse events
present, the greatest experience with this agent has been in patients with (anaphylaxis, aseptic meningitis, renal failure) related to IVIG therapy are
severe or refractory AD. fortunately uncommon. IgA deficiency should be suspected in individu-
als who develop anaphylaxis to IVIG.37 An explanation for why subjects
who have no IgA may develop anaphylaxis following IVIG infusion is
■ INTRAVENOUS
IMMUNOGLOBULIN ■ Table 94.4 Adverse effects potentially associated with IVIG

CLINICAL PHARMACOLOGY
Common
In healthy individuals, following IVIG infusion there is a biphasic
plasma IgG disappearance curve, with the initial phase representing Nausea
distribution between body compartments and early catabolism, and the Headache
second phase representing catabolism.29 The half-life of IVIG ranges Chills
from 14 to 24 days in healthy individuals and is more prolonged (26–35
Backache
days) in patients with humoral immunodeficiencies.29
Flushing

IMMUNOLOGIC EFFECTS Uncommon


IVIG has a variety of immunomodulating properties29 which are of po-
Renal insufficiency
tential benefit in immunologically mediated diseases such as immune
thrombocytopenic purpura and Kawasaki syndrome. These immunologic Aseptic meningitis
effects include blockade of Fc receptors, anti-cytokine effects, downregu- Anaphylaxis
lation of T- and B-cell function, inhibition of complement activation,

1648
Immunomodulators 94
suggested from the observation that IgA-deficient individuals may de- receiving IVIG was 16 mg/day, their mean FEV1 87% of predicted, and
velop anti-IgA IgE antibodies, as their immune system does not recognize their mean age 16, characteristics that were not significantly different
infused IgA as a self protein. In subjects who develop a severe headache from those of the placebo group. The primary outcome measured in the
related to IVIG therapy the development of aseptic meningitis should be study was the mean daily prednisone dose requirements (adjusted for
considered.38 These subjects will often have associated photophobia and a weight) determined during the observation month preceding the insti-
stiff neck, and their CSF will show a neutrophilic or a mixed cell pleocyto- tution of IVIG therapy, compared to the mean daily prednisone dose
sis.38 The induction of renal insufficiency by IVIG has also been reported requirements determined during month 7 of the study. The mean daily
and attributed to hyperosmolar renal damage induced by the breakdown prednisone dose requirements declined by 33% in the placebo group,
of sucrose, which is used as a stabilizer in IVIG preparations.39–41 and this reduction was not significantly different from the reduction in
As IVIG is derived from the pooled plasma of 3000–60 000 individu- prednisone in the 2 g/kg IVIG group (33% reduction in prednisone), or
als,36,42 the theoretic transmission of infectious agents requires contin- the 1 g/kg IVIG group (39% reduction in prednisone). There was also
ued vigilance. All IVIG donors are screened for HIV, hepatitis B, and no difference between the placebo and IVIG groups in other asthma
hepatitis C. To date there have been no reported cases of transmission of outcomes measured, including FEV1, emergency department visits,
HIV or hepatitis B following IVIG infusion.42 However, in the USA in hospitalizations, or missing days from work or school. The mean levels
1993–1994 there were outbreaks of hepatitis C associated with certain of IgE did not change significantly from baseline after the institution of
brands of IVIG, particularly those produced by chromatography.43–45 IVIG therapy. The study was terminated prematurely as 20% (3/15) of
Following the introduction of specific viral inactivation steps, no new the subjects randomized to the 2 g/kg IVIG group were hospitalized

DNA-BASED THERAPIES
cases of hepatitis C related to IVIG have been reported in the USA.46 with symptoms of headache, nausea, vomiting, photophobia, and men-
The theoretic potential for the transmission of Creutzfeld–Jakob disease ingismus, consistent with aseptic meningitis.34
(CJD) has also been reviewed by the FDA, NIH, and CDC, who suggest IVIG was also evaluated in a European double-blind placebo-
that the risk of transmission of CJD by blood products, if it exists, is con- controlled study in moderately severe asthmatics (mean FEV1 77% pre-
siderably lower than the risk for harm to public health from withdrawal of dicted) who had a mean age of 14 years.35 After a 2-month stabilization
the use of blood products.42,46 Although CJD transmission has never been phase, during which inhalation and oral corticosteroids were tapered to a
documented with transfusion of these products, the prolonged incubation minimum dose allowing effective control of asthma symptoms (mean dose
period and the lack of screening tests for CJD warrant continued research of inhaled corticosteroid of 1350 μg/day; 31% of subjects on oral corti-
and surveillance for the potential transmission of this rare condition. costeroids), subjects were randomized to receive IVIG (1g/kg) or placebo
monthly for 3 months. The addition of IVIG to a stable dose of inhaled
and/or oral corticosteroid did not result in a significant improvement in
CLINICAL STUDIES IN ASTHMA asthma symptoms, FEV1, or airway hyperreactivity to histamine at the
end of the study period. IVIG had no significant effect on IgE levels.
Open-label high-dose IVIG
In contrast to the open-label studies of IVIG, the double-blind
Three open-label studies in the USA of high-dose IVIG (2 g/kg) in placebo-controlled studies of IVIG34,35 do not demonstrate a significant
subjects with moderately severe asthma requiring daily or alternate-day improvement in asthma symptoms, emergency department visits, hospi-
prednisone have been performed.33,47,48 Overall, these studies demon- talizations, FEV1, airway hyperreactivity measurements, or reductions in
strate that IVIG (2 g/kg administered monthly for 6–9 months) has IgE levels. In addition, the high-dose IVIG in one study was associated
an oral corticosteroid-sparing effect, as in two studies subjects were with a significant risk for adverse events such as aseptic meningitis.
able to significantly reduce their daily oral corticosteroid dosage (from
21 mg to 10 mg48 and from 31 mg to 5 mg)47 or in one study their alter- CURRENT RECOMMENDATIONS
nate-day oral corticosteroid dosage (from 32 mg to 11 mg).33 Symptom
FOR USE IN CLINICAL PRACTICE
scores, glucocorticoid bursts for exacerbations of asthma, and hospitali-
zations for asthma were all significantly reduced in subjects who received Although the results of the open-label studies with IVIG in asthma
IVIG.33,47,48 No change in methacholine responsiveness or response to are encouraging in terms of their oral corticosteroid-sparing effect, the
exercise challenge was noted in subjects who received IVIG in these placebo-controlled studies do not confirm that IVIG has a corticoster-
studies.33,47,48 Although the open-label IVIG studies suggest that IVIG oid-sparing effect. The number of subjects randomized to IVIG groups
has a corticosteroid-sparing effect, the lack of a double-blind placebo- in the double-blind studies (9–16 subjects per IVIG group) are small,
controlled study design, and the small number of subjects studied (8–11 but similar numbers of subjects have been studied in the open-label
subjects in each study received IVIG)33,47,48 necessitate that these studies studies (8–11 subjects per IVIG group). At present IVIG is an experi-
be interpreted and validated in the context of additional double-blind mental therapy for asthma. It should be evaluated further in the context
placebo-controlled studies with IVIG which have been performed.34,35 of randomized, placebo-controlled clinical trials.

Double-blind studies of IVIG


Double-blind placebo-controlled studies of IVIG have been performed
■ DNA-BASED THERAPIES ■
in the USA and Europe.34,35 In the USA multicenter study,34 subjects Several types of DNA-based therapy are currently being evaluated in the
with corticosteroid-dependent asthma were randomized to receive treatment of allergy and asthma. Although these therapies are similar in
monthly infusions of either IVIG (2 g/kg or 1 g/kg) or placebo for that they all utilize oligodeoxynucleotides, they differ in structural DNA
7 months. At entry into the study the mean prednisone dose in the group sequences and their mechanism of action. The DNA-based therapies

1649
F THERAPEUTICS

are the predominant cells transfected after DNA inoculation via muscle
Promoter or skin injection, respectively. Cross-priming may serve as a mechanism
to augment or maintain the immune response.49 DNA vaccination has
generally been associated with the induction of a Th1 as opposed to a Th2
immune response. The presence of immunostimulatory DNA sequences
Antibiotic containing a CpG motif in the plasmid backbone of the DNA vaccine are
resistance considered to be important in generating the Th1 immune response.50

Antigen
Mouse models of allergy and asthma
coding The use of DNA vaccines encoding an allergen to modify the Th2 im-
cDNA mune response to that allergen has been studied extensively in mice.50
Both humoral and cellular immune responses to encoded allergens can
be generated following intramuscular or intradermal injection of plas-
mid DNA encoding a specific antigen.50 Furthermore, studies using a
CpG rat model of asthma have demonstrated that the IgE response, histamine
DNA-BASED THERAPIES

Plasmid backbone Transcription unit release into bronchoalveolar lavage fluid, and bronchial hyperreactivity
Th1 adjuvant Antigen synthesis following challenge with aerosolized dust mite allergen Der p5 were
inhibited in rats immunized with a plasmid DNA encoding the Der p 5
Fig. 94.4. Deoxyribonucleic acid (DNA) vaccine. DNA vaccines are allergen.51 The response to the DNA vaccine was antigen specific, as the
circular, extrachromosomal pieces of plasmid DNA that can be modified to
carry genes of interest in the antigen coding region (e.g. dust mite allergen rats immunized with plasmid DNA encoding the Der p 5 allergen were
for allergen immunotherapy). The transcription unit includes a promoter protected against Der p 5 allergen challenge, but not against a different
which initiates the transcription of the antigen coding sequence. The plasmid allergen, i.e., ovalbumin challenge.
backbone contains CpG (cytosine guanosine) motifs which bias the immune DNA vaccines also prevent anaphylactic reactions to peanut allergen (Ara
response to a Th1 response. Antibiotic resistance sequences allow for h 1) in mouse models of anaphylaxis.52 In these studies the DNA vaccine
selection of the plasmid during preparation of the DNA vaccine.
could be administered orally using chitin particles containing the DNA
plasmid encoding Ara h 1. The orally administered DNA vaccine remained
functional, as assessed by its ability to inhibit Th2 immune responses
currently being evaluated include DNA vaccines encoding the expression of as well as anaphylaxis following challenge with Ara h 1.52 Additional
allergens, antisense oligonucleotides which inhibit the translation of specific studies in mice have demonstrated that DNA vaccines can significantly
host cellular mRNA, and immunostimulatory DNA sequences containing reduce mortality associated with anaphylaxis.53 Moreover, these vaccines
a cytosine phosphorothioate-linked guanosine DNA motif (CpG DNA) can prevent allergic responses to birch pollen54 and latex55 allergens.
which bias the immune response away from a Th2 or pro-allergic response.
Potential use in human allergy and asthma
DNA VACCINES
DNA vaccines in humans have been evaluated in infectious disease and
DNA vaccines are circular, extrachromosomal pieces of plasmid DNA in oncology, in which no serious adverse events related to injection of
that can be modified to carry genes of interest (i.e. dust mite allergen for plasmid DNA have been reported.49 Although DNA vaccines alone
allergen immunotherapy)61 (Fig. 94.4). can elicit humoral and cellular immune responses to many antigens, the
immune response may be suboptimal for protection against infectious
disease.49 At present there are no published studies of the human im-
Immune response
mune response to DNA vaccines encoding allergens. Several potential
There are several potential mechanisms by which the antigen encoded by safety concerns regarding DNA vaccines will need to be addressed in
the injected plasmid DNA vaccine is processed and presented to elicit all studies, including the theoretical potential for integration of the in-
an immune response. The subcutaneously injected plasmid DNA could jected plasmid DNA into the host genome (with the theoretical risk of
be taken up by dendritic cells or somatic cells (skin and/or muscle) that increasing the risk for malignancy), as well as the potential for triggering
then transcribe and express the encoded antigen or allergen and elicit autoimmune responses to injected DNA. To date there is no evidence
an immune response.49 Alternatively, a non-antigen-presenting cell (i.e. that plasmids integrate into the host genome.49
muscle cell) injected with plasmid DNA could express and transfer the
protein (cross-priming) to a professional antigen-presenting cell.49 The
preponderance of evidence suggests that the primary immune response
after DNA vaccination is mediated by dendritic cells.49 During cross- IMMUNOSTIMULATORY DNA THERAPY
priming, antigen or peptides (both MHC class I and II) generated by
Immune response
plasmid DNA injected into somatic cells can be taken up by professional
antigen-presenting cells such as dendritic cells to prime T-cell responses. An alternative method of DNA-based immunization that has re-
Thus, somatic cells (i.e. myocytes and keratinocytes) may serve as a ceived significant attention recently is the use of CpG-rich (cytosine
significant reservoir of antigen available for cross-priming, as these cells phosphorothioate-linked guanosine DNA) immunostimulatory DNA

1650
Immunomodulators 94
A
Mouse models of allergy and asthma
5 Purine Purine C G Pyr Pyr 3
Several studies have demonstrated that CpG DNA can inhibit Th2
cytokine responses, as well as eosinophilic inflammation and airway hy-
CpG perreactivity to methacholine in mouse models of asthma.59 In addi-
tion to inhibiting eosinophilia of the airway and lung parenchyma, CpG
B DNA also significantly inhibits blood eosinophilia, suggesting that CpG
DNA exerts a significant effect on the bone marrow production and/or
CH3 the release of eosinophils.59 The inhibition of the bone marrow produc-
tion of eosinophils is associated with a significant inhibition of Th2 cell-
derived cytokine production (IL-4, IL-5).
The cellular mechanism of action of CpG DNA in vivo may be more
complex than that initially hypothesized based on results from in vitro
C G
studies. Although CpG DNA induces a strong Th1 response, the anti-
allergic effect of CpG DNA may not be mediated by this mechanism in
CpG vivo. In support of this hypothesis are studies demonstrating that adap-
tive transfer of antigen-specific Th1 cells does not protect against eosi-

DNA-BASED THERAPIES
Fig. 94.5. Cytosine–guanosine (CpG) DNA. A CpG DNA hexamer nophilic airway inflammation in a mouse model of asthma,60 and studies
sequences that have an optimal Th1 adjuvant effect in mice include the demonstrating that CpG DNA can mediate its anti-allergic effect in
motif 5′-purine–purine–CpG–pyrimidine–pyrimidine-3′, e.g. AACGTT or vivo independent of the Th1 cytokines interferon-γ and IL-12.61 Thus,
GACGTC. These sequences are non-coding. B Cytosine (C) methylation
(CH3) abolishes the Th1 adjuvant effect of CpG DNA sequences.
the ability of CpG DNA to induce a Th1 response and interferon-γ ex-
pression may not be essential to its inhibitory effect on allergic inflam-
mation in vivo.

Potential use in human allergy and asthma


sequences as inhibitors of Th2 responses to antigen. Interest in this
approach began when studies demonstrated that the CpG DNA motifs CpG-based therapeutics are currently undergoing evaluation in human
in Mycobacterium bovis BCG DNA induced interferon-γ, a cytokine clinical trials in allergy and asthma, infectious disease, and cancer.57 For
produced by Th1 cells.56 In contrast, the immunostimulatory effect of therapeutic use, CpG DNA oligodeoxynucleotides are typically syn-
vertebrate DNA is significantly lower than that of bacterial DNA. The thesized with a phosphorothiate linkage (rather than using the native
reduced immunostimulatory effect of vertebrate DNA is probably re- phosphodiester linkage) to prevent nuclease digestion and increase the
lated to a combination of the lower frequency of CpG DNA motifs in half-life of CpG. Subcutaneous administration of CpG DNA in healthy
vertebrate compared to bacterial DNA, as well as a high frequency of human volunteers results in high concentrations in the draining lymph
cytosine methylation in vertebrate compared to bacterial DNA, which node and induces high levels of serum cytokines.57 In contrast, intrave-
abolishes the immunostimulatory effect of vertebrate CpG DNA se- nous administration of the same CpG DNA results in a significant dilu-
quences.57 The DNA hexamer sequences generating the optimal Th1 tion in the blood, binding of CpG DNA to serum proteins, and failure
adjuvant effect in mice include the motif 5′-purine–purine–CpG– to induce a measurable serum cytokine response.57 Thus, the regional
pyrimidine–pyridimidine-3′, e.g. AACGTT or GACGTC (Fig. 94.5),57 lymph nodes play an important role in the immune response to subcu-
which activates the secretion of IFN-γ. CpG DNA has an indirect taneously administered CpG DNA. The most common side effect noted
effect on the adaptive immune response by activating the innate im- with subcutaneous injection of CpG DNA is a mild local injection site
mune system (e.g., plasmacytoid dendritic cells) to upregulate cytokine reaction. Some individuals may develop transient ‘flu-like’ symptoms of
expression (IL-12, IL-18, IFN-γ, IFN-α, IFN-β), upregulate MHC headache, fever, myalgia, and nausea.57 Although induction of autoim-
molecules, and upregulate co-stimulatory molecules.57 These effects on munity has not been noted, the duration of therapy with CpG DNA
the innate immune system lead to a cytokine milieu (IFN-γ+, IL-12+) in the majority of studies has been less than 6 months; hence further
which biases the T-lymphocyte immune response towards a Th1 re- studies are needed to address this issue.
sponse to newly encountered antigens. At present there are limited published results of CpG DNA in sub-
Studies have demonstrated an important role for a Toll receptor jects with allergy and asthma. In a phase I study of CpG DNA admin-
TLR9 (Toll receptor 9)58 in mediating signaling of CpG DNA, as the istered weekly by inhalation for 4 weeks to humans with mild asthma
deletion of genes encoding TLR-9 in mice results in mice that are un- (FEV1 87% of predicted), the nebulized CpG DNA induced expression
able to respond to CpG DNA.58 As TLR9 in humans is expressed pri- of IFN-γ in sputum cells, but did not attenuate the physiologic response
marily by plasmacytoid dendritic cells and B cells, CpG DNA exerts its to allergen inhalation challenge (early- or late-phase FEV1), sputum
immunomodulatory effects in humans predominantly through its effects eosinophils, or Th2 cytokines in sputum.62 Nebulized CpG DNA was
on plasmacytoid dendritic cells, which subsequently influence the adap- well tolerated and no significant adverse events were noted. Further
tive immune response. Toll-like receptors (expressed on a variety of in- studies are needed to determine whether different doses of CpG DNA,
nate immune cells) belong to a family of pattern recognition receptors different routes of CpG administration, or administration of CpG DNA
that are postulated to allow the innate immune system to sense different in clinical asthma (as opposed to a model of allergen-induced asthma),
classes of pathogen.57 demonstrate similar or different results.

1651
F THERAPEUTICS

ragweed protein CpG DNA conjugate has not been associated with
CYTOSINE PHOSPHOROTHIOATE GUANOSINE any significant side effects.64,65,67 The ability to reach a target dose of
12 μg of Amb a 1, the major ragweed allergen, without inducing a sys-
DNA CONJUGATED TO PROTEIN ALLERGEN
temic allergic reaction using only six escalating dose injections of the
Recently, interest has developed in the use of allergen protein conjugated conjugate is a significant advantage of the conjugate compared to stand-
to CpG DNA for the treatment of allergic diseases, as this approach has ard immunotherapy, which requires many more injections to safely reach
certain theoretical advantages compared to unconjugated CpG DNA. the maintenance dose. As some, but not all, studies have shown a clinical
The primary advantage of allergen protein conjugated CpG DNA is that benefit of the ragweed protein CpG DNA conjugate in subjects with
physically linking CpG DNA to antigens would increase the likelihood ragweed-induced allergic rhinitis, further large-scale studies are needed
of their delivery to the same antigen-presenting cell (APC), resulting in to determine both its effectiveness and its long-term safety.
an amplified, antigen-specific Th1 immune response,63 which is much less
likely to occur when CpG DNA and antigen are administered separately.
ANTISENSE OLIGODEOXYNUCLEOTIDE THERAPY
The same APC would therefore process the antigen and present the an-
tigen to Th cells, whereas CpG DNA would induce the APC to release The goal of antisense oligodeoxynucleotide (ODN) therapy is to selec-
IL-12, thus biasing the well-targeted Th cell to differentiate towards a tively inhibit the expression of a specific gene product by preventing the
Th1 phenotype. Furthermore, theoretically, the CpG DNA allergen pro- translation of mRNA into protein68 (Fig. 94.6). Antisense ODN therapy
DNA-BASED THERAPIES

tein conjugate might be less allergenic than the CpG DNA allergen pro- acts by sequence-specific hybridization to mRNA, inhibiting the expres-
tein mixture, as steric hindrance or electrostatic blockade of IgE-binding sion of that specific gene product while not affecting the expression of
epitopes could occur with CpG DNA coating the allergen.63 other genes. Antisense ODN therapy prevents the translation of the
RNA message into protein and promotes the degradation of the message
by ribonucleases. In order for antisense ODNs to be synthesized the cod-
Immune response
ing sequence of the gene to be inhibited must be known. Antisense com-
The immune response to Amb a 1, the major short ragweed allergen, pounds are generally about 20 base pairs of oligodeoxynucleotides that
conjugated to CpG DNA has been investigated in mice, rabbits, mon- have a sequence complementary to a portion of the targeted mRNA. The
keys, and humans.63 Administration of subcutaneous injections of a
ragweed protein CpG DNA conjugate to ragweed-allergic individuals
induced their peripheral blood mononuclear cells to express a Th1 rather Normal Antisense
than a Th2 cytokine profile.64 Individuals with ragweed-induced allergic DNA DNA
rhinitis who have received immunotherapy with a ragweed protein CpG
DNA conjugate respond to ragweed nasal challenge by increasing nasal
mucosa Th1 cytokine production, as well as by decreasing Th2 cytokine mRNA mRNA
production and nasal mucosa eosinophilia.65

Mouse models of allergy and asthma 5 3 mRNA


In a mouse model of asthma, concomitant administration of antigen 3 5 Antisense
conjugated to CpG DNA intratracheally prior to allergen challenge re-
duced airway eosinophilia and airway hyperreactivity to methacholine,
downregulated the Th2 immune response, and enhanced Th1 cytokine
expression.66 The allergen protein conjugated CpG DNA was found to
be 100 times more efficient than the unconjugated mixture in reducing
airway eosinophilia and bronchial hyperreactivity, with this inhibitory 5 3 mRNA
effect observed for at least 2 months. CpG DNA allergen protein con-
jugates therefore offer promise as therapy for asthma and other allergic
disorders, although investigation of its immune modulating properties in
humans requires further study. Protein mRNA
degradaiton

Potential use in human allergy and asthma


Reduced translation
In human studies a ragweed protein CpG DNA conjugate induces sig- of mRNA to protein
nificantly less basophil histamine release than does ragweed alone.63 In Fig. 94.6. Antisense oligodeoxynucleotides. Transcription of genes
a phase II clinical study of 25 individuals with allergic rhinitis due to follows the sequence of DNA, mRNA, and translation into protein.
ragweed, a short-course of immunotherapy with six weekly injections of Antisense oligonucleotides are designed to have complementary sequences
a ragweed protein CpG DNA conjugate significantly reduced rhinitis to target and bind to specific known mRNA sequences. This binding blocks
symptom scores during the ragweed season, and these symptom improve- access of the mRNA/antisense hybrid to the ribosome and translation of the
mRNA to protein. An enzyme Rnase H degrades the RNA strand of the
ments persisted through the second ragweed season without additional
mRNA/antisense hybrid.
immunotherapy.67 Other than mild local injection site reactions the

1652
Immunomodulators 94
poor stability of oligonucleotides in vivo has been improved by modify- lead to a new class of drugs that switch off unwanted expression of
ing the phosphodiester backbone to a sulfur-containing phosphorothio- individual genes in disease.72 Two gene-silencing drugs using the RNA
ate backbone, which enhances the stability of oligonucleotides to resist interference strategy have been designed to treat macular degeneration
enzymatic degradation. and are already in clinical trials.72
Post-transcriptional inhibition of gene expression at the mRNA level
can therefore be accomplished not only by antisense-based therapy but
Animal models of allergy and asthma
also by small interfering RNAs (siRNAs).72 siRNAs are small double-
Several gene products important to asthma and allergic inflammation stranded RNAs (about 21 nucleotides) designed to have complemen-
have been targeted with antisense ODN in animal models of asthma. tarity to a specific single-stranded mRNA, and mediate destruction
These include cell surface receptors (adenosine A1, IL-5 receptor), cy- of the specific target mRNA.72 A pivotal point in mRNA silencing is
tokines (IL-4, stem cell factor), intracellular signaling molecules (Syk the selection of the active strand of the siRNA duplex. The translation
protein tyrosine kinases), and transcription factors (GATA-3).68–70 of the ability of siRNA to silence genes in cell culture in vitro and in
The feasibility of using antisense therapy in asthma was first sug- animal models in vivo to human application still requires significant
gested in studies using antisense targeted to the adenosine A1 receptor further development. In particular, improving targeting siRNAs in vivo
in a rabbit model of asthma.68 Pretreatment of rabbits with inhaled without their degradation, as well as assessing the risks and benefits of
adenosine A1 receptor antisense resulted in an increase in the dose of such an approach in humans, remains problematic. siRNAs have been
aerosolized adenosine required to reduce the dynamic compliance of delivered by inhalation to the airway in mice and shown to inhibit genes

MYCOBACTERIAL VACCINES
the lung (a measure of bronchoconstriction) by 50%.68 Airway smooth such as heme oxygenase-1 in the lung, but not in other organs.73 Phase I
muscle derived from rabbits treated with inhaled adenosine A1 re- human studies have begun to evaluate siRNA in the treatment of ocular
ceptor antisense demonstrated an approximately 75% decrease in the neovascularization,72 but no human studies have yet been reported in
adenosine A1 receptor density, but did not affect the adenosine A2 allergy or asthma.
receptor density, indicating the specificity of the targeted antisense
therapy.68
Potential use in human allergy and asthma
GATA-3 is a transcription factor which is preferentially expressed
in Th2 cells and regulates the expression of Th2 cytokine genes im- The potential for antisense to be utilized in human disease has best been
portant to allergic inflammation. Studies with antisense targeted to applied to cytomegalovirus (CMV) infection in subjects with AIDS.
GATA-3 in mouse models of asthma have demonstrated that GATA-3 Vitravene is an antisense therapy that inhibits CMV replication and
antisense inhibits eosinophilic airway inflammation, airway hyperre- is approved for local therapy of CMV retinitis in patients with AIDS,
activity, and cytokine expression, including IL-4 and IL-5.70 Target- providing conceptual proof of principle that antisense therapy can be
ing a transcription factor that regulates the expression of several Th2 used to treat human disease.74 At present there is no published infor-
cytokine genes is theoretically advantageous over targeting a single mation on the use of antisense therapy in the treatment of human al-
cytokine, although this approach has the potential for increased side lergic disease or asthma.
effects.
Interestingly, antisense therapy to ameliorate allergic inflammation
has also been targeted at grass pollen allergens to reduce the generation
of pollen as opposed to targeting the allergic patient. The potential of
■ MYCOBACTERIAL VACCINES ■
this approach has been demonstrated in studies of antisense-mediated Epidemiologic studies suggest an inverse correlation between rates
silencing of a gene encoding Lol p 5, a major ryegrass pollen allergen.71 of tuberculosis and the prevalence of asthma in children. In Japanese
A pollen-specific promoter was used to drive the antisense expression schoolchildren the rate of current symptoms of asthma was reduced by
of Lol p 5 in ryegrass plants. Approximately two-thirds of the IgE re- approximately one-third in positive tuberculin responders. Based on
activity of ryegrass pollen has been attributed to Lol p 5. The transgenic epidemiologic evidence that mycobacterial exposure inhibits the devel-
ryegrass pollen showed significantly reduced allergenicity, as reflected opment of allergy and asthma, animal model and subsequent clinical
by low IgE-binding capacity of the transgenic pollen compared to that studies with heat-killed Mycobacterium vaccae or heat-inactivated bacil-
of control pollen.71 The transgenic ryegrass plants in which Lol p 5 lus Calmette–Guérin (BCG) have been investigated as a potential im-
gene expression is perturbed showed normal fertile pollen development, munomodulatory therapy for asthma and allergy.
indicating that genetic engineering of hypoallergenic grass plants is
possible.
ANIMAL MODELS OF ALLERGY AND ASTHMA
ALTERNATIVES TO ANTISENSE: SMALL Intranasal infection of mice with Mycobacterium bovis–BCG 1–3 months
prior to allergen challenge significantly inhibits the development of air-
INTERFERING RNAs (siRNA)
way eosinophilia in a mouse model of allergen-induced asthma.75 The
In 2006 the Nobel Prize in Physiology or Medicine was awarded to two mycobacterium-induced inhibition of airway eosinophila is associated
scientists, Andrew Fire and Craig Mello, for their far-reaching discovery with inhibition of the Th2 cytokine IL-5, but not with inhibition of
in 1998 about how genes are controlled within living cells. They discov- IgE. The mycobacterium-induced inhibition of airway eosinophilia was
ered an unexpected system of gene regulation in living cells that resulted significantly reduced in interferon-γ receptor-deficient mice, suggesting
in a subsequent explosive phase of research in a field known variously as an important role for the Th1 cytokine IFN-γ in mediating the anti-
RNA interference or gene silencing. RNA interference may potentially eosinophilic effect of the mycobacterial infection.

1653
F THERAPEUTICS

POTENTIAL USE IN HUMAN ALLERGY have also been shown to be highly effective in treating a number of other
systemic inflammatory autoimmune diseases, including ankylosing
AND ASTHMA
spondylitis, psoriatic arthritis, psoriasis, and Crohn’s disease.79
Two different Mycobacterium vaccae vaccines (heat-killed or delipidated The success of TNF-α inhibitors in RA represents a proof-of-concept
deglycolipidated) have been investigated in a double-blind study of that inhibition of a single cytokine can be effective in treating a dis-
moderately severe atopic asthmatics (mean FEV1 76%; mean inhaled ease in which multiple cytokines are expressed, if that cytokine serves
corticosteroid dose 500–600 μg of inhaled beclomethasone daily) with a key or central role. Most RA patients respond to TNF-α inhibitors
stable symptoms.76 Outcomes measured included the effect on asthma with a reduction in signs and symptoms, improved quality of life, and
severity (change in morning peak flow) and immunologic response preservation of functional status.79 Radiographs demonstrate objective
3 months later. This study demonstrated that there was no clinical benefit evidence that TNF-α inhibitors also significantly slow disease progres-
or immunologic difference (eosinophil, IgE, T-cell proliferation, cytokine sion in RA to an extent not seen with any previous agents. Interestingly,
response) in subjects who received two intradermal doses of either of although initial experience with TNF inhibitors was most commonly in
the two Mycobacterium vaccae vaccines tested compared to those who re- RA patients with chronic refractory disease, more recent studies have
ceived placebo.76 Additional ongoing studies with Mycobacterium vaccae demonstrated even better clinical outcomes when TNF inhibitors were
in asthma and allergy will determine whether the lack of benefit noted used earlier in the disease course.80 As TNF inhibitors do not induce
in this particular study relates to a lack of effectiveness of Mycobacterium immunologic tolerance, maintenance of clinical efficacy almost always
CYTOKINE-BASED THERAPY

vaccae immunization, or alternatively, relates to study design issues (route requires continued therapy, certainly for patients with long-standing RA.
of administration, study population, outcome measure, sample size, etc). Importantly, 2–4-year and longer studies of anti-TNF-α therapy in RA
suggest that the clinical response is well maintained. Nevertheless, some
HEAT-INACTIVATED BCG THERAPY IN HUMAN patients fail to respond to anti-TNF therapy and others eventually lose
their response. Interestingly, patients failing one TNF inhibitor may re-
ALLERGY AND ASTHMA
spond to therapy with another.
The effect on asthma severity of administering four weekly injections Inhibition of TNF has also been studied in patients with asthma,81–84
of heat-inactivated BCG has been investigated in a placebo-controlled although results have not been as clear cut nor as dramatic in all asthma
study of 3 months’ duration in moderately severe asthmatics who were studies as in autoimmune conditions. Examples of studies in asthma
Mantoux skin test negative.77 The heat-inactivated BCG did not im- which have shown a benefit to TNF blockade include studies of the
prove any of the markers of asthma severity (FEV1, peak flow, asthma soluble TNF receptor etanercept in 10 patients with severe asthma, which
exacerbations, asthma symptom scores, β-agonist use), and also had noted improvements in FEV1, airway responsiveness to methacholine,
no effect on blood eosinophil and serum IgE levels. Recruitment for and asthma-related quality of life.81 In another study of 38 symptomatic
the trial was halted early and the number of injections was reduced in subjects with moderate asthma, infliximab did not demonstrate significant
some patients owing to excessive local injection site reactions to BCG.77 clinical efficacy in terms of the primary endpoint of lung function.83 How-
In addition to the lack of efficacy of repeated heat-inactivated BCG in- ever, there was a significant reduction in the number of moderate asthma
jections, the occurrence of severe local injection site reactions limits the exacerbations in the infliximab group compared to placebo.83 It is possible
therapeutic application of this approach in asthma. that only a subset of severe asthmatics respond to anti-TNF therapy, and
that levels of expression of membrane-bound TNF-α by peripheral blood
monocytes in such subjects with asthma may predict responses.81
■ CYTOKINE-BASED THERAPY ■
IL-4
Cytokines play a key role in regulating the initiation, perpetuation,
and resolution of allergic inflammation (Chapter 10). Based on stud- IL-4 mediates several important proinflammatory functions in allergic
ies demonstrating that several cytokines are expressed in the airway in inflammation, including induction of the IgE isotype switch, induction of
asthmatics,78 novel therapeutics have been developed to target individual vascular cell adhesion molecule-1, promotion of eosinophil transmigration
cytokines in patients with asthma to determine whether any of these across the endothelium, stimulation of mucus production, and promotion
individual cytokines may play an important role in disease pathogenesis. of Th2 lymphocyte differentiation. The therapeutic potential of a recom-
Pre-clinical studies in animal models of asthma have demonstrated that binant soluble IL-4 receptor (IL-4R) as an IL-4 antagonist has been stud-
targeting individual cytokines such as TNF, IL-4, or IL-5 reduces levels ied in asthmatics. In two small studies, treatment with the IL-4 receptor
of eosinophilic airway inflammation and airway responsiveness. antagonist improved asthma symptom scores and pulmonary function,
reduced β2-agonist rescue use, as well as lowering levels of exhaled nitric
oxide.85,86 However, subsequently two large phase III studies in moderate-
TNF
to-severe asthma failed to reveal efficacy, possibly due to dose limitations
To date, the principle that one can target a single cytokine and have a and the short duration of action of the IL-4 receptor antagonist.
significant impact on human disease expression has been best validated
in the treatment of autoimmune disease such as rheumatoid arthritis
IL-5
(RA) with inhibitors of the cytokine TNF-α.79 Currently there are three
TNF inhibitors used in clinical practice: infliximab (a chimeric mono- As IL-5 is a key regulator of eosinophil proliferation, studies have inves-
clonal antibody), etanercept (a soluble TNF receptor/Fc fusion protein), tigated whether targeting IL-5 would reduce eosinophilic inflammation
and adalimumab (a human monoclonal antibody).79 TNF inhibitors and improve asthma outcomes. In asthmatics, therapy with an anti-IL-5

1654
Immunomodulators 94
antibody has a dramatic effect in reducing blood eosinophilia (~ 90%), 16. Armstrong V, Oellerich M. New developments in the immunosuppressive drug monitoring of
cyclosporin, tacrolimus, and azathioprine. Clin Biochem 2001; 34:9.
but interestingly does not inhibit the late-phase lower airway response 17. Fonacier L, Spergel J, Charlesworth EN, et al. Report of the Topical Calcineurin Inhibitor
to inhaled allergen,87 nor improve measures of clinical asthma,88 thereby Task Force of the American College of Allergy Asthma and Immunology and the American
Academy of Allergy Asthma and Immunology. J Allergy Clin Immunol 2005; 115:1249.
raising questions about the previously held notion of the pivotal roles of 18. Schreiber S, Crabtree G. The mechanism of action of ciclosporin A and FK506. Immunol Today
IL-5 and eosinophils in allergic asthma. Subsequent studies have dem- 1992; 13:136.
19. Vanrenterghem Y. Which calcineurin inhibitor is preferred in renal transplantation: tacrolimus
onstrated that anti-IL-5 is less effective at inhibiting eosinophils in the or cyclosporin? Curr Opin Nephrol Hypertens 1999; 8:669.
lower airways (~50%) than in the blood (~90%), indicating that partial 20. Van Den Borne B, Landewe R, Houkes I, et al. No increased risk of malignancies and
mortality in cyclosporin A treated patients with rheumatoid arthritis. Arthritis Rheum 1998;
depletion of tissue eosinophils might be insufficient for clinical efficacy 41:1930.
in a complex disease such as asthma. Interestingly, anti-IL-5 reduces 21. Alexander A, Barnes N, Kay AB. Trial of cyclosporin in corticosteroid-dependent chronic severe
asthma. Lancet 1992; 339:324.
levels of airway remodeling in asthma through inhibition of eosinophil 22. Lock S, Kay AB, Barnes N. Double blind placebo controlled study of cyclosporin A as a
expression of TGF-β,89 and improves eosnophilia and clinical responses corticosteroid-sparing agent in corticosteroid dependent asthma. Am J Respir Crit Care Med
1996; 153:509.
in patients with eosinophilic esophagitis.90 23. Khan L, Kon O, MacFarlane A, et al. Attenuation of the allergen-induced late asthmatic
reaction by cyclosporin A is associated with inhibition of bronchial eosinophils, interleukin-5,
granulocyte macrophage colony-stimulating factor, and eotaxin. Am J Respir Crit Care Med
2000; 162:1377.
24. Nizankowska E, Soja J, Pinis G, et al. Treatment of steroid-dependent bronchial asthma with
cyclosporin. Eur Respir J 1995; 8:1091.
■ CONCLUSIONS ■ 25. Sowden J, Berth-Jones J, Ross J, et al. Double-blind, controlled, crossover study of cyclosporin in
adults with severe refractory atopic dermatitis. Lancet 1991; 338:137.

REFERENCES
26. De Rie M, Meinardi M, Dos J. Lack of efficacy of topical cyclosporin A in atopic dermatitis and
Immunomodulatory therapy has made a significant therapeutic impact allergic contact dermatitis. Acta Dermatol Venereol 1991; 71:452.
in immunologically mediated diseases such as rheumatoid arthritis, but 27. Reitamo S. Tacrolimus: A new topical immunomodulatory therapy for atopic dermatitis.
J Allergy Clin Immunol 2001; 107:445.
the search for safe and effective immunomodulator therapy contin- 28. Feutren G, Mihatsch M. Risk factors for cyclosporin-induced nephropathy in patients with
ues in immunologically mediated diseases such as asthma and allergy. autoimmune diseases. N Engl J Med 1992; 326:1654.
At present, however, immunomodulator therapy (other than traditional
allergen protein immunotherapy and anti-IgE) remains investigational Intravenous immunoglobulin
in the field of asthma and allergic disorders. 29. Morell A. Pharmacokinetics of intravenous immunoglobulin preparations. In: Lee ML, Strand
V, eds. Intravenous immunoglobulins in clinical practice. New York: Marcel Dekker, 1997.
30. Yu Z, Lennon VA. Mechanism of intravenous immune globulin therapy in antibody-mediated
autoimmune disease. N Engl J Med 1999; 340:227–228.
31. Sigman K, Ghibu F, Sommerville W, et al. Intravenous immunoglobulin inhibits IgE
References production in human B lymphocytes. J Allergy Clin Immunol 1998; 102:421.
32. Ballow M. Immunology: is steroid-dependent asthma a disease treatable with intravenous
immunoglobulin? Clin Immunol 1999; 91:123.
Methotrexate 33. Mazer BD, Gelfand EW. An open-label study of high-dose intravenous immunoglobulin in
severe childhood asthma. J. Allergy Clin Immunol 1991; 87:976.
1. Cronstein B. Molecular therapeutics: methotrexate and its mechanism of action. Arthritis
34. Kishiyama JL, Valacer D, Cunningham-Rundles C, et al. A multicenter, randomized, double-
Rheum 1996; 39:1951.
blind, placebo-controlled trial of high-dose intravenous immunoglobulin for oral corticosteroid-
2. Ortiz Z, Shea B, Suarez-Almazor M, et al. Folic acid and folinic acid for reducing side effects
dependent asthma. Clin Immunol 1999; 91:126.
in patients receiving methotrexate for rheumatoid arthritis. Cochrane Database Syst Rev 2000;
35. Niggemann B, Leupold W, Schuster A, et al. Prospective, double-blind, placebo-controlled,
CD000951.
multicentre study on the effect of high-dose, intravenous immunoglobulin in children and
3. Genestier L, Paillot R, Fournel S, et al. Immuno-suppressive properties of methotrexate:
adolescents with severe bronchial asthma. Allergy 1998; 28:205.
apoptosis and clonal deletion of activated peripheral T cells. J Clin Invest 1998; 102:322.
36. Orange JS, Hossny EM, Weiler CR, et al. Use of intravenous immunoglobulin in human disease:
4. Cronstein B, Eberle M, Gruber H, et al. Methotrexate inhibits neutrophil function by stimulating
a review of evidence by members of the Primary Immunodeficiency Committee of the American
adenosine release from connective tissue cells. Proc Natl Acad Sci USA 1991; 88:2441.
Academy of Allergy, Asthma and Immunology. J Allergy Clin Immunol 2006; 117:S525,.
5. Kremer J, Alarcon G, Lightfoot R, et al. Methotrexate for rheumatoid arthritis: suggested
37. Burks AW, Sampson HA, Buckley RH. Anaphylactic reactions after gamma globulin
guidelines for monitoring liver toxicity. Arthritis Rheum 1994; 37:316.
administration in patients with hypogammaglobulinemia. N Engl J Med 1986; 314:560.
6. Chalmers RLG, Kirby B, Smith A, et al. Replacement of routine liver biopsy by procollagen
38. Sekul EA, Cupler EJ, Dalakas MC. Aseptic meningitis associated with high-dose intravenous
III aminopeptide for monitoring patients with psoriasis receiving long-term methotrexate:
immunoglobulin therapy: frequency and risk factors. Ann Intern Med 1994; 121:259.
a multicentre audit and health economic analysis. Br J Dermatol 2005; 152:444.
39. Ahsan N. Intravenous immunoglobulin induced nephropathy: a complication of IVIG therapy.
7. Kremer J, Alarcon G, Weinblatt M, et al. Clinical, laboratory, radiographic and histopathologic
J Nephrol 1998; 11:157.
features of methotrexate-associated lung injury in patients with rheumatoid arthritis. Arthritis
40. Stahl M, Schifferli JA. The renal risks of high-dose intravenous immunoglobulin treatment.
Rheum 1997; 40:1829.
Nephrol Dial Transplant 1998; 13:2182.
8. Mullarkey M, Blumenstein B, Andrade W, et al. Methotrexate in the treatment of corticosteroid-
41. Center for Disease Control and Prevention. Renal insufficiency and failure associated with
dependent asthma: a double-blind crossover study. N Engl J Med 1988; 318:603.
immune globulin intravenous therapy – United States, 1985–1998. MMWR Morb Mortal
9. Shiner RJ, Nunn AJ, Chung KF, et al. Trial of methotrexate in steroid dependent asthma. Lancet
Wkly Rep 1999; 48:518.
1990; 336:137.
42. Lederman HM. Intravenous immunoglobulin and specific antibody therapies for
10. Erzurum S, Leff J, Cochran J, et al. Lack of benefit of methotrexate in severe, steroid-dependent
immunodeficiency and autoimmune/inflammatory disorders. Allergy: Principle and Practice
asthma: a double-blind, placebo-controlled study. Ann Intern Med 1991; 114:353.
Updates 2001; 1:1.
11. Guss S, Portnoy J. Methotrexate treatment of severe asthma in children. Pediatrics 1992;
43. Bresee JS, Mast EE, Coleman PJ, et al. Hepatitis C virus infection associated with administration
89:635.
of intravenous immune globulin. JAMA 1996; 276:1563.
12. Marin M. Low-dose methotrexate spares steroid usage in steroid-dependent asthmatic patients.
44. Rossi G, Tucci A, Cariani E, et al. Outbreak of hepatitis C virus infection in patients with
A meta-analysis. Chest 1997; 112:29.
hematologic disorders treated with intravenous immunoglobulins: different prognosis according
13. Davies H, Olson L, Gipson P. Methotrexate as a steroid sparing agent for asthma in adults.
to the immune status. Blood 1997; 90:1309.
Cochrane Database Syst Rev 2 2000; CD000391.
45. Kenny-Walsh E. Clinical outcomes after hepatitis C infection from contaminated anti-D
immune globulin. N Engl J Med 1999; 340:1228.
46. Center for Disease Control and Prevention. Availability of immune globulin intravenous for
Immunophilin-binding agents and calcineurin inhibitors: cyclosporin, treatment of immune deficient patients – United States, 1997–1999. MMWR Morb Mortal
tacrolimus, and pimecrolimus Wkly Rep 1999; 48:159.
47. Landwehr LP, Jeppson JD, Katlan MG, et al. Benefits of high-dose iv immunoglobulin in
14. Sigal N, Dumont F. Cyclosporin A, FK-506, and rapamycin: pharmacologic probes of patients with severe steroid-dependent asthma: Chest 1998; 114:1349.
lymphocyte signal transduction. Annu Rev Immunol 1992; 10:519. 48. Spahn JD, Leung DYM, Chan MTS, et al. Mechanisms of glucocorticoid reduction in
15. Yocum D. Cyclosporin, FK-506, rapamycin and other immunomodulators. Rheum Dis Clin asthmatic subjects treated with intravenous immunoglobulin: J Allergy Clin Immunol 1999;
North Am 1996; 22:133. 103:421.

1655
F THERAPEUTICS

DNA-based therapies 71. Bhalla PL, Swoboda I, Singh MB. Antisense-mediated silencing of a gene encoding a major
ryegrass pollen allergen. Proc Natl Acad Sci USA 1999; 96:11676.
49. Gurunathan S, Klinman D, Seder RA. DNA vaccines: immunology, application, and 72. Xie FY, Woodle MC, Lu PY. Harnessing in vivo siRNA delivery for drug discovery and
optimization. Annu Rev Immunol 2000; 18:927. therapeutic development. Drug Discov Today 2006; 11:67.
50. Raz E, Tighe H, Sato Y, et al. Preferential induction of a Th1 immune response and inhibition 73. Zhang X, Shan P, Jiang D, et al. Small interfering RNA targeting heme oxygenase-1 enhances
of specific IgE antibody formation by plasmid DNA immunization. Proc Natl Acad Sci USA ischemia–reperfusion-induced lung apoptosis. J Biol Chem 2004; 279:10677.
1996; 93:5141. 74. Galderisi U, Cascino A, Giordano A. Antisense oligonucleotides as therapeutic agents. J Cell
51. Hsu CH, Chua KY, Tao MH, et al. Immunoprophylaxis of allergen-induced immunoglobulin Physiol 1999; 181:251.
E synthesis and airway hyperresponsiveness in vivo by genetic immunization. Nat Med 1996;
2:540.
52. Roy K, Mao HQ, Huang SK, et al. Oral gene delivery with chitosan – DNA nanoparticles Mycobacterial vaccines
generate immunologic protection in a murine model of peanut allergy. Nat Med 1999; 5:387.
53. Horner AA, Nguyen MD, Ronaghy A, et al. DNA-based vaccination reduces the risk of lethal 75. Erb KJ, Holloway JW, Sobeck A, et al. Infection of mice with Mycobacterium bovis–bacillus
anaphylactic hypersensitivity in mice. J Allergy Clin Immunol 2000; 106:349. Calmette–Guérin (BCG) suppresses allergen induced airway eosinophilia. J Exp Med 1998;
54. Hartl A, Kiesslich J, Weiss R, et al. Immune response after immunization with plasmid DNA 187:561.
encoding Bet v 1, the major allergen of birch pollen. J Allergy Clin Immunol 1999; 103:107. 76. Shirtcliffe PM, Easthope SE, Cheng S, et al. The effect of delipidated deglycolipidated
55. Slater JE, Paupore E, Zhang YT, et al. The latex allergen Hev b 5 transcript is widely distributed (DDMV) and heat-killed Mycobacterium vaccae in asthma. Am J Respir Crit Care Med 2001;
after subcutaneous injection in BALB/c mice of its DNA vaccine. J Allergy Clin Immunol 1998; 163:1410.
102:469. 77. Shirtcliffe PM, Easthope SE, Weatherall M, Beasley R. Effect of repeated intradermal injections
56. Tokunaga T, Yamamoto H, Shimada S, et al. Antitumor activity of deoxyribonucleic acid of heat-inactivated Mycobacterium bovis–bacillus Calmette–Guérin in adult asthma. Clin Exp
fraction from Mycobacterium bovis BCG.I. Isolation, physicochemical characterization, and Allergy 2004; 34:207.
antitumor activity. J Natl Cancer Inst 1984; 72:955.
57. Krieg AM. Therapeutic potential of Toll-like receptor 9 activation. Nature Rev Drug Discov
2006; 5:471.
Cytokine-based therapy
REFERENCES

58. Hemmi H, Takeuchi O, Kawal T, et al. A Toll-like receptor recognizes bacterial DNA. Nature
2000; 408:740. 78. Broide DH, Lotz M, Cuomo AJ, et al. Cytokines in symptomatic asthma airways. J Allergy Clin
59. Broide D, Schwarze J, Tighe H, et al. Immunostimulatory DNA sequences inhibit IL-5, Immunol 1992; 89:958.
eosinophilic inflammation, and airway hyperresponsiveness in mice. J Immunol 1998; 161:7054. 79. Kavanaugh A, Broide DH. Cytokine/chemokine antagonists. In: Lichtenstein LL, Busse
60. Hansen G, Berry G, DeKruyff RH, et al. Allergen-specific Th1 cells fail to counterbalance Th2 WW, Geha RS, eds. Current therapy in allergy immunology and rheumatology, 6th edn.
cell-induced airway hyperreactivity but cause severe airway inflammation. J Clin Invest 1999; Philadelphia: Mosby, 2003:432–435.
103:175. 80. Taylor PC, Steuer A, Gruber J, et al. Ultrasonographic and radiographic results from a two-
61. Kline JN, Krieg AM, Waldschmidt TJ, et al. CpG oligodeoxynucleotides do not require Th1 year controlled trial of immediate or one-year-delayed addition of infliximab to ongoing
cytokines to prevent eosinophilic airway inflammation in a murine model of asthma. J Allergy methotrexate therapy in patients with erosive early rheumatoid arthritis. Arthritis Rheum
Clin Immunol 1999; 104:1258. 2006; 54:47.
62. Gauvreau GM, Hessel EM, Boulet LP, et al. Immunostimulatory sequences regulate interferon- 81. Berry MA, Hargadon B, Shelley M, et al. Evidence of a role of tumor necrosis factor alpha in
inducible genes but not allergic airway responses. Am J Respir Crit Care Med 2006; 174:12. refractory asthma. N Engl J Med 2006; 354:697.
63. Tighe H, Takabayashi K, Schwartz D, et al. Conjugation of immunostimulatory DNA to the 82. Erzurum SC. Inhibition of tumor necrosis factor alpha for refractory asthma. N Engl J Med
short ragweed allergen amb a 1 enhances its immunogenicity and reduces its allergenicity. 2006; 354:754.
J Allergy Clin Immunol 2000; 106:124. 83. Erin EM, Leaker BR, Nicholson GC, et al. The effects of mAb directed against tumor necrosis
64. Simons FE, Shikishima Y, Van Nest G, et al. Selective immune redirection in humans with factor-alpha in asthma. Am J Respir Crit Care Med 2006; 174:753.
ragweed allergy by injecting Amb a 1 linked to immunostimulatory DNA. J Allergy Clin 84. Howarth PH, Babu KS, Arshad HS, et al. Tumour necrosis factor (TNFalpha) as a novel
Immunol 2004; 113:1144. therapeutic target in symptomatic corticosteroid dependent asthma. Thorax 2005; 60:1012.
65. Tulic MK, Fiset PO, Christodoulopoulos P, et al. Amb a 1-immunostimulatory 85. Borish LC, Nelson HS, Corren JC, et al. Efficacy of soluble IL-4 receptor for the treatment if
oligodeoxynucleotide conjugate immunotherapy decreases the nasal inflammatory response. adults with asthma. J Allergy Clin Immunol 2001; 107:963.
J Allergy Clin Immunol 2004; 113:235. 86. Borrish LC, Nelson HS, Lanz MJ, et al. Interleukin-4 receptor in moderate atopic asthma.
66. Shirota H, Sano K, Kikuchi T, et al. Regulation of murine airway eosinophilia and Th2 cells A phase 1/randomized, placebo-controlled trial. Am J Respir Crit Care Med 1999; 160:1816.
by antigen-conjugated CpG oligodeoxynucleotides as a novel antigen specific modulator. 87. Leckie MJ, ten Brinke A, Khan J, et al. Effects of an interleukin-5 blocking monoclonal
J Immunol 2000; 164:5575. antibody on eosinophils, airway hyperresponsiveness and the late asthmatic response. Lancet
67. Creticos P, Schroeder J, Hamilton R, et al. Immunotherapy with a ragweed-Toll-like receptor 2000; 356:2144.
9 agonist vaccine for allergic rhinitis. N Engl J Med 2006; 355:1445. 88. Kips JC, O’Connor BJ, Langley SJ, et al. Effect of SECH 55700, a humanized anti-human
68. Richardson PJ. Asthma. Blocking adenosine with antisense. Nature 1997; 385:684. interleukin-5 antibody in severe persistent asthma. Am J Respir Crit Care Med 2003;
69. Lach-Trifilieff E, McKay RA, Monia BP, et al. In vitro and in vivo inhibition of interleukin 167:1655.
(IL)-5-mediated eosinopoiesis by murine IL-5R alpha antisense oligonucleotide. Am J Respir 89. Flood-Page P, Menzies-Gow A, Phipps S, et al. Anti-IL-5 treatment reduces deposition of
Cell Mol Biol 2001; 24:116. ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics.
70. Inotto S, De Sanctis GT, Lehr HA, et al. Treatment of allergic airway inflammation and J Clin Invest 2003; 112:1029.
hyperresponsiveness by antisense-induced local blockade of GATA-3 expression. J Exp Med 90. Stein ML, Collins MH, Villanueva JM, et al. Anti-IL-5 (mepolizumab) therapy for eosinophilic
2001; 193:1247. esophagitis. J Allergy Clin Immunol 2006; 118:1312.

1656

You might also like