You are on page 1of 5

Autoimmunity Reviews 8 (2009) 682–686

Contents lists available at ScienceDirect

Autoimmunity Reviews
j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / a u t r ev

Infection and type 1 diabetes mellitus — A two edged sword?


Elad Goldberg a, Ilan Krause b,c,⁎
a
Infectious Diseases Unit, Rabin Medical Center, Beilinson Hospital, Israel
b
Department of Medicine E, Rabin Medical Center, Beilinson Hospital, Israel
c
Sackler Faculty of Medicine, Tel-Aviv University, Israel

a r t i c l e i n f o a b s t r a c t

Article history: Infection by various viral and bacterial pathogens has long been proposed as one of the
Received 23 January 2009 etiologies of autoimmune diabetes. Many theories, ranging from direct cytolysis of pancreatic
Accepted 9 February 2009 islet cells to immunological processes such as antigen mimicry and polyclonal lymphocyte
Available online 13 February 2009
activation, tried to explain the epidemiological correlation between infections and diabetes,
supported by information from human and animal studies. However, a direct correlation and
Keywords:
exact mechanism continue to elude investigators due to scarce and conflicting data.
Type 1 diabetes
Infections
Interestingly, there is also data to support an opposite role for infection in the development
Viruses of type 1 diabetes, as several pathogens demonstrated a protective effect from this disease. This
Bacteria article reviews the current data available from clinical studies and animal models, while trying
Autoimmunity to explain the different mechanisms underlying these findings.
© 2009 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 682
2. Infection as a possible trigger of T1DM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 683
2.1. Viruses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 683
2.1.1. Enteroviruses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 683
2.1.2. Cytomegalovirus (CMV) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 683
2.1.3. Other viruses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 683
2.2. Other pathogens. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 684
3. Infections protecting from type 1 DM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 684
4. Comments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 685
Take-home messages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 685
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 685

1. Introduction environmental triggers involved in the pathogenesis of the


disease are yet to be determined. Identifying these triggers
The autoimmune etiology of type 1 diabetes mellitus has become of utmost importance as the incidence of T1DM is
(T1DM) has been studied extensively for many years. The increasing [2]. Different environmental factors are implicated
immunological mechanisms leading to the destruction of in the etiology of autoimmune disease, and among them the
pancreatic β-cells are clearer today, as are the genetic factors role of infection is a long standing assumption studied by
responsible for host susceptibility [1]. However, the exact numerous investigators [3]. Three main mechanisms explain-
ing this relation were proposed: activation of polyclonal
⁎ Corresponding author. Department of Medicine E, Rabin Medical Center,
lymphocytes by the infection, protein sequences of the
Beilinson Campus, Petach Tikva 49100, Israel. infecting pathogen mimicking autoantigens, and increased
E-mail address: ikrause@post.tau.ac.il (I. Krause). immunogenicity due to target organ inflammation, inducing

1568-9972/$ – see front matter © 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.autrev.2009.02.017
E. Goldberg, I. Krause / Autoimmunity Reviews 8 (2009) 682–686 683

an autoimmune response. All three mechanisms result in the identify viral infection in the pancreatic tissue of T1DM
induction of autoantibodies and other processes recognized patients. Indirect effects of viral infections on various
in the pathogenesis of autoimmune diseases [4]. However, it immunological mechanisms have also been described for
should be noted that direct causal relation between an different viruses in humans and animals.
infecting agent and an autoimmune disease has not yet
been solidly confirmed. 2.1.1. Enteroviruses
Interestingly, research of an opposite relation between First evidence for the relation between enteroviruses and
infection and autoimmune diseases has gained momentum. T1DM relied on antibody assays, demonstrating increased
This originated in the “hygiene theory”, noting that allergic prevalence of enteroviral IgM in the sera of T1DM patients
and autoimmune diseases increased in frequency in the [10] and in mothers to children with T1DM compared with
developed countries while infectious diseases became rarer, controls [11], although not all reports reproduced these
suggesting a protective effect of infection in relation to findings. Subsequently, these findings were strengthened by
autoimmune diseases [5]. Most of the data on this relation newer molecular methods which detected enteroviral RNA in
comes from epidemiological studies correlating the presence the blood of T1DM patients. However, the attempts to
of antibodies to certain pathogens and the incidence of determine the exact mechanism underlying the correlation
autoimmune disease [6]. Further data is available from animal between enteroviruses and T1DM met with difficulties.
models. Different enteroviruses are capable of directly destroying
Infection as an environmental trigger for T1DM has been human pancreatic islet cells in vivo. However, it was shown in
studied extensively, especially in laboratory setting and animal models that infection of the cells may be dependent
animal models, less so in clinical trials. Data on the effect of on the presence of pre-existing inflammation [12]. Further-
different infections on β-cells and the development of more, genetic factors also affect the ability of enteroviruses
autoimmune diabetes is growing, but no major conclusions and other viruses to damage islet cells and induce diabetes
have been made yet. In this review we will attempt to [13]. Other studies in animal models did not demonstrate a
organize and present the existing data and discuss its direct effect of enteroviruses on islet cells, but an indirect
implications. autoimmune response probably induced by the virus [14].
Therefore, enteroviruses may play a significant role in the
2. Infection as a possible trigger of T1DM development of T1DM in association with immunologic and
genetic host factors.
As specified above, the role of infection in the develop-
ment of T1DM has been reported both in animal models and 2.1.2. Cytomegalovirus (CMV)
in humans. As aforementioned, 3 major mechanisms were CMV has been implicated as a trigger for the development
suggested as an explanation for this role: of T1DM in several case reports and following demonstration
of a correlation between viral genome identified in leucocytes
1) Activation of polyclonal B or T cells by the infecting agent,
and autoantibodies to islet and β-cells in T1DM patients [15].
thereby triggering production of different autoantibodies,
However, several other reports cast doubt on this relation, as
including islet-cell antibodies [7]. Data supporting this
positive serology for CMV was not associated with develop-
mechanism is, however, scarce and inconclusive.
ment of T1DM or of β-cell autoantibodies. Animal models
2) Mimicking of autoantigens by pathogen proteins. This
support the role of CMV in the development of T1DM, either
mechanism has been implicated in autoimmune diseases
through activation of macrophages and proliferation of
such as multiple sclerosis and Guillaine Barré syndrome. It
autoreactive T-cells [16] or through induction of islet-cell
was also proposed for T1DM, with viral proteins mimick-
specific autoantibodies [17]. The search for the exact
ing autoantigens such as GAD65 and inducing T-cell
mechanism involving CMV infection and T1DM is still under
reaction [8]. Attempts to reproduce this reaction were
investigation both in clinical and laboratory studies. More
not always successful, casting doubt on this mechanism.
recently, the effect of CMV on different complications in solid
3) Infection induced inflammation increasing the expression
organ transplanted patients, including post-transplantation
of molecules which participate in the process of autoanti-
diabetes, has been studied extensively. The incidence of new
gen recognition by T-cells. This phenomenon was sup-
onset diabetes is higher among transplanted patients with
ported in several animal model studies [9], but was not
asymptomatic CMV infection [18], but the exact role of the
directly demonstrated in human studies.
virus in this relation is still investigated.
The bulk of the data in T1DM patients concerns different viral
infections, while other pathogens are reported sporadically. 2.1.3. Other viruses
Several other viruses were investigated as potential
2.1. Viruses triggers for T1DM. Among the most recently investigated are
rotaviruses, a major cause of diarrhea among the pediatric
Viral infections have been implicated in the pathogenesis population. Epidemiological data demonstrated a correlation
of T1DM as early as the 1970s. Several theories of pathogen- between rotavirus infection and first appearance of antibodies
esis were raised, according to the different type of virus and to insulin and islet-cell antigens [19]. It should be noted that
its effects on pancreatic tissue and the immune system. One this correlation was not observed in other reports, specifically
such theory is that of direct infection and lysis of pancreatic β- in T1DM genetically susceptible children [20]. Results from
cells. This is supported by different case reports in humans, several studies in animal models remain inconclusive; how-
and in animal models as well. However, other reports failed to ever it appears that timing of the infection in relation to
684 E. Goldberg, I. Krause / Autoimmunity Reviews 8 (2009) 682–686

existing inflammation may determine the effect of the virus on controls [31]. Other anecdotal reports exist for bacterial
the development of T1DM [21]. pathogens such as pertussis and even protozoal pathogens
Two other viruses implicated as causing T1DM are rubella such as schistosoma, but none of them has so far been
and mumps. Epidemiological data demonstrated an increased established.
rate of T1DM among children with congenital rubella or
mumps infection. Furthermore, increase in the incidence of 3. Infections protecting from type 1 DM
autoimmune diabetes appeared to have been restrained
following the introduction of the measles–mumps–rubella As mentioned before, there is a correlation between a
(MMR) vaccine [22]. However, the incidence of T1DM decrease in infectious diseases in the developed countries and
resumed its upward trend since then. Both viruses can infect a continuous rise in the incidence of T1DM. This is supported
human β-cells, but do not induce their destruction [23,24]. by further epidemiological data demonstrating higher rate of
Rubella is known to mimic antigens recognized by T-cells of T1DM in low-crowded households [32] and in first born
diabetic patients [25], while mumps is involved in cytokine children (less exposed to infections in their siblings).
release from human β-cells [26]. Although it is unclear Supporting evidence is available from animal models. It has
whether these viruses play a significant role in the pathogen- been shown that diabetes may be prevented in susceptible
esis of T1DM, it appears they are involved in immunologic laboratory animals by introduction of mycobacterial cell wall
processes related to this disease. components [33].
Other viruses have been reported as possible cause for Animal models have been helpful in understanding the
T1DM (parvovirus, encephalomyocarditis virus, influenza, underlying processes of this protective effect. Two proposed
retroviruses), but data on these pathogens is scant, and no explanations for this effect are homeostatic competition
pathogenetic mechanisms have been established for any of (infection induced lymphocyte expansion and competition
them. with other lymphocytes involved in the autoimmune process)
and bystander suppression (suppressive immune response to
2.2. Other pathogens infection affects the autoimmune process).
A recently investigated mechanism is the effect of Toll like
Bacterial infections, although less than viral, have been receptors (TLRs) on activation of the immune system and
investigated as possible triggers for autoimmune diseases, possible regulation of autoimmune responses. TLRs are a
including T1DM. Bacterial DNA is known to stimulate the group of receptors recognizing different microbial and fungal
immune system to produce DNA autoantibodies that may ligands and participating in the immune system response to
play a role in autoimmune diseases [27]. When addressing infections. It has been shown that TLRs are major contributors
T1DM, the possible mechanisms underlying the effect of to the protective effect of infection on the development of
bacteria on the pancreas resemble the ones proposed for diabetes, as shown in diabetes susceptible animals receiving
viruses (i.e., direct damage to β-cells, immunological bacterial extracts.
mechanisms such as antigen mimicry, activation of lympho- These findings further clarify the complexity and diversity
cytes and modification of cytokine activity). A bacterial of the effects of infection on autoimmune responses and
mechanism irrelevant to viruses is toxin mediated damage diseases, specifically T1DM.
to the pancreas. This has been observed in animal models One of the first evidence for bacterial protection against
using Streptomyces species and the bafilomycin toxin they T1DM was the observation that BCG vaccination (prepared
produce. The toxin caused glucose intolerance, reduction in from Mycobacterium bovis) of NOD mice suppresses overt
islet size and β-cells mass [28]. It also caused disruption of the diabetes [34]. This observation was repeated with M. avium,
pancreatic organogenesis in utero, thus accelerating the onset which prevented diabetes in NOD mice, although the animals
of diabetes [29]. However, these findings were not consoli- were resistant to the infection [35]. Another interesting
dated by additional data. pathogen which was implicated as a cause for T1DM, but
More recently, newer mechanisms of bacterial involve- prevents the disease in animal models is Schistosoma mansoni.
ment in T1DM were proposed and investigated. Alterations in It appears that exposure to antigens of the Schistosoma worm
the intestinal microbiota have been shown to affect the or eggs modulates immune responses of T-cells and inter-
development of diabetes in animal models, and administra- leukin levels, and thereby prevents T1DM in NOD mice [36].
tion of different antimicrobial agents decreased the incidence The most explored protective pathogen in animal models is
of T1DM in these animals [30]. There is increasing data that the lymphocytic choriomeningitis virus (LCMV), which
the gut flora is involved in different immunological processes, prevents the development of T1DM in NOD mice. When
including autoimmunity. Different factors, such as the timing infected with this virus, the mice did not develop lymphocytic
of introduction of different agents to the gut flora and a infiltrates of the pancreatic islets (which did appear in
possible leak that triggers immunological and autoimmune uninfected mice). Furthermore, lymphocytes from infected
responses, have been proposed as explanations for this animals also prevented T1DM in uninfected mice [37]. This
phenomenon. However, this mechanism remains elusive as model has been duplicated in other trials, but clinical data in
some of the microbial agents induce protection from diabetes humans is lacking. Several other pathogens demonstrated a
while others may trigger the disease. protective effect in animals susceptible to diabetes. Salmo-
Other bacterial pathogens were also implicated in the nellatyphimurium infection has been shown to prevent T1DM
development of T1DM. One of them is Mycobacterium in NOD mice, although the protective mechanism is not clear
avium, whose DNA was detected in the peripheral blood of [38]. Even the Coxsackie virus which was extensively
a large proportion of T1DM patients compared with healthy investigated as a culprit in T1DM, was shown to produce
E. Goldberg, I. Krause / Autoimmunity Reviews 8 (2009) 682–686 685

long term protection from the disease in NOD mice when the [7] Schatz DA, Barrett DJ, Maclaren NK, Riley WJ. Polyclonal nature of islet cell
antibodies in insulin-dependent diabetes. Autoimmunity 1988;1:45–50.
infection precedes the inflammatory process [39]. Another [8] Hiemstra HS, Schloot NC, van Veelen PA, Willemen SJ, Franken KL, van
virus implicated as a trigger for T1DM, Rotavirus, was also Rood JJ, et al. Cytomegalovirus in autoimmunity: T cell crossreactivity to
shown to provide protection from development of T1DM in viral antigen and autoantigen glutamic acid decarboxylase. Proc Natl
Acad Sci U S A 2001;98:3988–91.
infant and young adult NOD mice following oral and intra- [9] Horwitz MS, Bradley LM, Harbertson J, Krahl T, Lee J, Sarvetnick N.
peritoneal administration [40]. Diabetes induced by Coxsackie virus: initiation by bystander damage
and not molecular mimicry. Nat Med 1998;4:781–5.
[10] King ML, Shaikh A, Bidwell D, Voller A, Banatvala JE. Coxsackie-B-virus-
4. Comments specific IgM responses in children with insulin-dependent (juvenile-
onset; type I) diabetes mellitus. Lancet 1983;1:1397–9.
The relation between infection and T1DM has been [11] Hyoty H, Hiltunen M, Knip M, Laakkonen M, Vahasalo P, Karjalainen J, et al.
A prospective study of the role of Coxsackie B and other enterovirus
studied for more than 4 decades. Data has been accumulating
infections in the pathogenesis of IDDM. Childhood Diabetes in Finland
both from human studies and animal studies, shedding light (DiMe) Study Group. Diabetes 1995;44:652–7.
on the different pathogens involved in the etiology of diabetes [12] Drescher KM, Kono K, Bopegamage S, Carson SD, Tracy S. Coxsackievirus
and the different mechanisms responsible for this relation. As B3 infection and type 1 diabetes development in NOD mice: insulitis
determines susceptibility of pancreatic islets to virus infection. Virology
the amount of data increases, more pathogens are identified 2004;329:381–94.
and additional mechanisms are proposed. However, direct [13] Flodstrom M, Tsai D, Fine C, Maday A, Sarvetnick N. Diabetogenic
data linking these pathogens to T1DM is scarce, and there are potential of human pathogens uncovered in experimentally permissive
beta-cells. Diabetes 2003;52:2025–34.
conflicting findings that do not enable a full understanding of [14] Horwitz MS, Ilic A, Fine C, Balasa B, Sarvetnick N. Coxsackieviral-
the biological pathways behind this correlation. Even more mediated diabetes: induction requires antigen-presenting cells and is
baffling is the fact that several pathogens, including ones that accompanied by phagocytosis of beta cells. Clin Immunol 2004;110:
134–44.
are implicated as causing T1DM, seem to provide protection [15] Pak CY, Eun HM, McArthur RG, Yoon JW. Association of cytomegalovirus
from this disease under certain conditions. infection with autoimmune type 1 diabetes. Lancet 1988;2:1–4.
It is obvious that infectious pathogens, especially viruses, [16] van der Werf N, Hillebrands JL, Klatter FA, Bos I, Bruggeman CA, Rozing J.
Cytomegalovirus infection modulates cellular immunity in an experimen-
are involved in biological and immunological processes that tal model for autoimmune diabetes. Clin Dev Immunol 2003;10:153–60.
contribute to the development of autoimmune diabetes. It is [17] Pak CY, Cha CY, Rajotte RV, McArthur RG, Yoon JW. Human pancreatic
also clear that these processes are complex and not yet fully islet cell specific 38 kilodalton autoantigen identified by cytomegalovirus-
induced monoclonal islet cell autoantibody. Diabetologia 1990;33:569–72.
understood. In order to clarify the role of infection in the
[18] Hjelmesaeth J, Sagedal S, Hartmann A, Rollag H, Egeland T, Hagen M, et al.
etiology of diabetes, both animal and human studies are direly Asymptomatic cytomegalovirus infection is associated with increased risk
required, so that we may keep up with the ever changing of new-onset diabetes mellitus and impaired insulin release after renal
epidemiological picture of infection and autoimmunity. transplantation. Diabetologia 2004;47:1550–6.
[19] Honeyman MC, Coulson BS, Stone NL, Gellert SA, Goldwater PN, Steele
CE, et al. Association between rotavirus infection and pancreatic islet
Take-home messages autoimmunity in children at risk of developing type 1 diabetes. Diabetes
2000;49:1319–24.
[20] Blomqvist M, Juhela S, Erkkila S, Korhonen S, Simell T, Kupila A, et al.
• Epidemiological and laboratory findings support the corre- Rotavirus infections and development of diabetes-associated autoanti-
lation between infections and type 1 diabetes. bodies during the first 2 years of life. Clin Exp Immunol 2002;128:511–5.
• Different viruses, especially enteroviruses, and bacteria are [21] Graham KL, Sanders N, Tan Y, Allison J, Kay TW, Coulson BS. Rotavirus
infection accelerates type 1 diabetes in mice with established insulitis.
involved in the etiology of autoimmune diabetes through J Virol 2008;83:6139–49.
immunological processes and direct damage to the pancreas. [22] Hyoty H, Hiltunen M, Reunanen A, Leinikki P, Vesikari T, Lounamaa R,
• Infectious agents may also be associated with protection et al. Decline of mumps antibodies in type 1 (insulin-dependent)
diabetic children and a plateau in the rising incidence of type 1
against the development of autoimmune diabetes. diabetes after introduction of the mumps–measles–rubella vaccine in
• The timing of the infection and the type of pathogen may Finland. Childhood Diabetes in Finland Study Group. Diabetologia
determine the different effect on the immune system, thus 1993;36:1303–8.
[23] Numazaki K, Goldman H, Seemayer TA, Wong I, Wainberg MA. Infection
triggering the disease or protecting the host from it.
by human cytomegalovirus and rubella virus of cultured human fetal
• The exact mechanisms behind these findings remain islets of Langerhans. In Vivo 1990;4:49–54.
elusive, and necessitate further research. [24] Vuorinen T, Nikolakaros G, Simell O, Hyypia T, Vainionpaa R. Mumps and
Coxsackie B3 virus infection of human fetal pancreatic islet-like cell
clusters. Pancreas 1992;7:460–4.
References [25] Ou D, Mitchell LA, Metzger DL, Gillam S, Tingle AJ. Cross-reactive rubella
virus and glutamic acid decarboxylase (65 and 67) protein determinants
[1] Shoenfeld Y, Gilburd B, Abu-Shakra M, Amital H, Barzilai O, Berkun Y, et al. recognised by T cells of patients with type I diabetes mellitus. Diabetologia
The mosaic of autoimmunity: genetic factors involved in autoimmune 2000;43:750–62.
diseases—2008. Isr Med Assoc J 2008;10:3–7. [26] Cavallo MG, Baroni MG, Toto A, Gearing AJ, Forsey T, Andreani D, et al.
[2] Incidence and trends of childhood type 1 diabetes worldwide 1990– Viral infection induces cytokine release by beta islet cells. Immunology
1999. Diabet Med 2006;23:857–66. 1992;75:664–8.
[3] Shoenfeld Y, Zandman-Goddard G, Stojanovich L, Cutolo M, Amital H, [27] Pisetsky DS. The role of innate immunity in the induction of autoimmunity.
Levy Y, et al. The mosaic of autoimmunity: hormonal and environmental Autoimmun Rev 2008;8:69–72.
factors involved in autoimmune diseases—2008. Isr Med Assoc J 2008;10: [28] Myers MA, Hettiarachchi KD, Ludeman JP, Wilson AJ, Wilson CR, Zimmet
8–12. PZ. Dietary microbial toxins and type 1 diabetes. Ann N Y Acad Sci
[4] Shoenfeld Y, Blank M, Abu-Shakra M, Amital H, Barzilai O, Berkun Y, et al. 2003;1005:418–22.
The mosaic of autoimmunity: prediction, autoantibodies, and therapy in [29] Hettiarachchi KD, Zimmet PZ, Myers MA. Transplacental exposure to
autoimmune diseases—2008. Isr Med Assoc J 2008;10:13–9. bafilomycin disrupts pancreatic islet organogenesis and accelerates
[5] Bach JF. The effect of infections on susceptibility to autoimmune and diabetes onset in NOD mice. J Autoimmun 2004;22:287–96.
allergic diseases. N Engl J Med 2002;347:911–20. [30] Brugman S, Klatter FA, Visser JT, Wildeboer-Veloo AC, Harmsen HJ,
[6] Ram M, Anaya JM, Barzilai O, Izhaky D, Porat Katz BS, Blank M, et al. The Rozing J, et al. Antibiotic treatment partially protects against type 1
putative protective role of hepatitis B virus (HBV) infection from diabetes in the bio-breeding diabetes-prone rat. Is the gut flora involved
autoimmune disorders. Autoimmun Rev 2008;7:621–5. in the development of type 1 diabetes? Diabetologia 2006;49:2105–8.
686 E. Goldberg, I. Krause / Autoimmunity Reviews 8 (2009) 682–686

[31] Sechi LA, Paccagnini D, Salza S, Pacifico A, Ahmed N, Zanetti S. Myco- immune response and prevent onset of type 1 diabetes. Eur J Immunol
bacterium avium subspecies paratuberculosis bacteremia in type 1 2003;33:1439–49.
diabetes mellitus: an infectious trigger? Clin Infect Dis 2008;46:148–9. [37] Oldstone MB. Viruses as therapeutic agents. I. Treatment of nonobese
[32] Patterson CC, Carson DJ, Hadden DR. Epidemiology of childhood IDDM insulin-dependent diabetes mice with virus prevents insulin-depen-
in Northern Ireland 1989–1994: low incidence in areas with highest dent diabetes mellitus while maintaining general immune competence.
population density and most household crowding. Northern Ireland J Exp Med 1990;171:2077–89.
Diabetes Study Group. Diabetologia 1996;39:1063–9. [38] Zaccone P, Raine T, Sidobre S, Kronenberg M, Mastroeni P, Cooke A.
[33] Sadelain MW, Qin HY, Lauzon J, Singh B. Prevention of type I diabetes in Salmonella typhimurium infection halts development of type 1 diabetes
NOD mice by adjuvant immunotherapy. Diabetes 1990;39:583–9. in NOD mice. Eur J Immunol 2004;34:3246–56.
[34] Harada M, Kishimoto Y, Makino S. Prevention of overt diabetes and [39] Serreze DV, Wasserfall C, Ottendorfer EW, Stalvey M, Pierce MA, Gauntt C,
insulitis in NOD mice by a single BCG vaccination. Diabetes Res Clin et al. Diabetes acceleration or prevention by a Coxsackievirus B4 infection:
Pract 1990;8:85–9. critical requirements for both interleukin-4 and gamma interferon. J Virol
[35] Bras A, Aguas AP. Diabetes-prone NOD mice are resistant to Mycobac- 2005;79:1045–52.
terium avium and the infection prevents autoimmune disease. Immu- [40] Graham KL, O'Donnell JA, Tan Y, Sanders N, Carrington EM, Allison J, et al.
nology 1996;89:20–5. Rotavirus infection of infant and young adult nonobese diabetic mice
[36] Zaccone P, Fehervari Z, Jones FM, Sidobre S, Kronenberg M, Dunne DW, involves extraintestinal spread and delays diabetes onset. J Virol
et al. Schistosoma mansoni antigens modulate the activity of the innate 2007;81:6446–58.

Evaluation of the efficacy and safety of pamapimod, a p38 MAP kinase inhibitor, in a double-blind, methotrexate-controlled
study of patients with active rheumatoid arthritis

New therapies are under development to improve the efficacy in the treatment of rheumatoid arthritis, monotheparies should
promise to be good enough as traditional ones in the improvement of disease course and symptoms. In a recent study, Cohen SB
et al, (Arthritis and rheumatism 2009;60:335–344) intended to compare the efficacy and safety of pamapimod (a selective
inhibitor of the alpha-isoform of p38 MAP kinase) as monotherapy in comparison with methotrexate (MTX) treatment in adult
patients with active rheumatoid arthritis (RA). Patients in a randomly assigned to 1 of 4 treatment groups and received 12 weeks of
double-blind treatment. One group received MTX (7.5 mg/week with planned escalation to 20 mg/week), and 3 groups received
pamapimod (50, 150, or 300 mg) once daily. The primary efficacy end point was the proportion of patients meeting the American
College of Rheumatology 20% improvement criteria (achieving an ACR20 response) at 12 weeks. Secondary end points included
ACR50 and ACR70 responses, change from baseline in the Disease Activity Score in 28 joints (DAS28), categorical analyses of
DAS28/European League Against Rheumatism response, and change from baseline in each parameter of the ACR core set of
measures. Safety monitoring included recording of adverse events (AEs), laboratory testing, immunology assessments, adminis-
tration of electrocardiograms, and assessment of vital signs. As result, patients assigned to receive MTX and pamapimod had similar
demographics and baseline characteristics. At week 12, fewer patients taking pamapimod had an ACR20 response (23%, 18%, and
31% in the 50-, 150-, and 300-mg groups, respectively) compared with patients taking MTX (45%). Secondary efficacy end points
showed a similar pattern. AEs were typically characterized as mild and included infections, skin disorders, and dizziness.
Pamapimod was generally well tolerated, but the 300-mg dose appeared to be more toxic than either the 2 lower doses or MTX. The
researchers concluded that pamapimod was not as effective as MTX in the treatment of active RA.

Monoarticular corticosteroid injection versus systemic administration in the treatment of rheumatoid arthritis patients: a
randomized double-blind controlled study

Monoarticular corticosteroid injection versus systemic administration in the treatment of rheumatoid arthritis patients is still an
interest are for disccusion. Konai MS, et al. (Clin Exp Rheumatol 2009;27:214–21) intended to compare the efficacy and safety of
intraarticular glucocorticoid injection to its systemic use for treatment of knee synovitis in rheumatoid patients. A randomized
double-blind controlled study was conducted including 60 patients with RA. Patients were randomized to receive either a single
intraarticular knee injection with triamcinolone hexacetonide 60 mg (3 ml) and xylocaine chloride 2% (1 ml) associated to a single
intramuscular injection of 1 ml of xylocaine chloride 2% (IAI group) or 1 ml of xylocaine chloride 2% by intraarticular injection and a
intramuscular injection of triamcinolone acetonide 60 mg (3 ml) and xylocaine chloride 2% (1 ml) (IM group). All patients were
blindfolded for the procedure. Evaluations were performed at baseline and 1, 4, 8 and 12 weeks post-intervention. The following
instruments were used: VAS for knee pain, as primary outcome, VAS for knee morning stiffness and edema; the ACR 20, 50 and 70%
improvement criteria; knee circumference and goniometry; Likert's scale of improvement; daily use of oral glucocorticoid and
NSAIDs, blood pressure and adverse effects. The researchers found that patients in the IAI group had significantly better results for
VAS for knee pain, edema and morning stiffness as well as for improvement evaluation after intervention according to the patient
(pb 0.001) and physician (p= 0.02). The results demonstrate that intraarticular injection with glucocorticoids is superior to its
systemic use for the management of monoarticular synovitis in rheumatoid patients. The intraarticular approach showed better
results in terms of local inflammatory variables and improvement evaluation by the patient and physician.

You might also like