Skip to main content
Proceedings of the American Thoracic Society logoLink to Proceedings of the American Thoracic Society
. 2007 Jul;4(3):289–294. doi: 10.1513/pats.200701-021AW

Antiinfective Applications of Toll-like Receptor 9 Agonists

Arthur M Krieg 1
PMCID: PMC2647632  PMID: 17607015

Abstract

The innate immune system detects pathogens by the presence of highly conserved pathogen-expressed molecules, which trigger host immune defenses. Toll-like receptor (TLR) 9 detects unmethylated CpG dinucleotides in bacterial or viral DNA, and can be stimulated for therapeutic applications with synthetic oligodeoxynucleotides containing immune stimulatory “CpG motifs.” TLR9 activation induces both innate and adaptive immunity. The TLR9-induced innate immune activation can be applied in the prevention or treatment of infectious diseases, and the adaptive immune–enhancing effects can be harnessed for improving vaccines. This article highlights the current understanding of the mechanism of action of CpG oligodeoxynucleotides, and provides an overview of the preclinical data and early human clinical trial results, applying these TLR9 agonists in the field of infectious diseases.

Keywords: innate immunity, dendritic cell, infection, cytokine


The family of Toll-like receptors (TLRs) appears to play a pivotal role in the innate immune system for the detection of highly conserved, pathogen-expressed molecules. To enable the rapid detection of infection, each of the 10 TLRs currently known to be expressed in humans has apparently evolved to be stimulated in the presence of certain types of pathogen-expressed molecules, which are either not expressed in host cells, or are sequestered in cellular compartments where they are unavailable to the TLRs. Activation of a TLR by an appropriate pathogen molecule acts as an “alarm signal” for initiation of the appropriate immune defenses.

A central task of the innate immune system may well be to determine whether a newly encountered pathogen is present extracellularly, as with most bacteria and fungi, in which case the “correct” type of immune response to be initiated is a T helper cell (Th) 2–like response, or intracellularly, as with most viruses and intracellular bacteria, in which case the innate immune system should induce a Th1-like immune response capable of killing infected cells. Recent studies indicate that the innate immune system accomplishes this feat, at least in part, by making use of the ligand specificity and the cellular site of expression of the TLRs (1). This review focuses on TLR9, which is expressed within the endosomal compartments of some innate immune cells, where it appears to function as a sensor for intracellular infection (2).

Biologists most commonly think of DNA as the genetic blueprint of life. The therapeutic applications of DNA, such as gene therapy, antisense, and DNA vaccines, have primarily made use of mechanisms for manipulating gene expression based on Watson-Crick base pairing. However, in recent years it has become clear that the immune system has evolved defense mechanisms against infections that are based on the detection of a subtle difference in the chemical structure of host DNA from that of viruses and bacteria. The innate immune system appears to use TLR9 for detecting unmethylated CpG dinucleotides, which are relatively common in bacterial and viral genomes, but are highly methylated and uncommon in vertebrate genomes (3). Bacterial and many viral DNAs generally contain the expected random frequency of about 1 CpG dinucleotide per 16 bases, but CpG dinucleotides are markedly suppressed in vertebrate genomes to about 25% of the expected frequency if base utilization was random. In addition, and in contrast to the unmethylated CpG dinucleotides in viral or bacterial DNAs, vertebrate genomes are highly CpG methylated: approximately 80% of the time that a C is followed by a G, the C is methylated at the 5′ position, but C in other positions is not preferentially methylated.

TLR9 activation by CpG DNA or synthetic oligodeoxynucleotides (ODNs) induces strong Th1-like immune activation, with the secretion of type-I IFN and activation of natural killer (NK) cells and strong CD8+ T-cell responses (reviewed in Reference 4). The targeting of TLR9 has emerged as a powerful tool in the generation of Th1 adaptive immunity, and has shown promise for enhancing the efficacy of vaccination.

THE ROLE OF TLR9 IN THE MECHANISM OF ACTION OF CpG ODNs

A critical advance in understanding the mechanism of action of the CpG motif was the identification of its receptor, TLR9 (3). Including TLR9, 10 human TLRs have been identified to date, and function as one family of what have been termed “pattern recognition receptors” (PRRs) (reviewed in Reference 2). PRRs have a general ability to detect certain molecular structures that are conserved in certain pathogens, but are not present or are not accessible to the appropriate PRRs in self tissues. Other examples of TLR ligands include certain lipopeptides (detected by TLR2), double-stranded RNA (TLR3), endotoxins (TLR4), and flagellin (TLR5). The immune system appears to use the presence of any of these molecular structures as a “danger signal” that indicates the presence of one general type of infection and activates appropriate defense pathways.

Among resting human immune cells, TLR9 is expressed primarily or exclusively in B cells and in plasmacytoid dendritic cells (pDCs) (5). In some studies, functional TLR9 expression has been reported in activated human neutrophils, monocytes and monocyte-derived cells, activated CD4 T cells, pulmonary epithelial cells, NK cells, and intestinal epithelium, but in other studies this has not been observed, or the cells showed no direct response to CpG ODNs (reviewed in Reference 4).

Unfortunately, the cellular patterns of TLR expression vary between different species, so the results of TLR stimulation in one species may not be predictive of what will occur in another. For example, mice differ from primates in that they express TLR9 not only in pDCs and B cells, but also in monocytes and myeloid DC. This makes it difficult at best to use observations with CpG ODNs in murine studies to predict accurately the effects of TLR9 activation in humans.

Administration of a CpG ODN activates pDCs to secrete IFN-α in both rodents and humans, promoting Th1 adaptive immune responses. TLR9-stimulated B cells and pDCs show increased expression of costimulatory molecules, resistance to apoptosis, up-regulation of the chemokine receptor, CCR7, and secretion of Th1-promoting chemokines and cytokines, such as macrophage inflammatory protein-1, IFN-γ–inducible protein-10, and other IFN-inducible genes (6). These effects drive the migration and clustering of pDCs in the T-cell regions of lymph nodes and other lymphoid tissues. Coactivation of naive, germinal center, or memory B cells through the B-cell antigen receptor and TLR9, can be strong enough to drive their differentiation into antibody-secreting plasma cells (7). In the case of memory B cells, which have been stimulated previously, activation through TLR9 alone is sufficient to drive differentiation to plasma cells (8, 9). The magnitude of this B-cell–stimulatory effect is greater than that of any other single B-cell mitogen, and has provided applications for CpG ODNs in the production of antigen-specific human antibodies. The efficiency of hybridoma generation from purified primary human memory B cells is improved from 1 to 2% without a CpG ODN, to 30 to 100% with the addition of PF-3512676 (formerly known as CPG 7909, or ODN 2006) (10). CpG-induced plasma cell differentiation does not require T-cell help, but its efficiency is enhanced further by interactions with pDCs and by B-cell receptor cross-linking (11). The molecular signaling pathways downstream from TLR9 have recently been reviewed in detail, and are discussed further here (2). The net effect of TLR9 activation is to induce Th1-biased cellular and humoral effector functions of innate and adaptive immunity.

There are at least three distinct classes of immune stimulatory CpG ODNs: A, B, and C classes (4). A-class CpG ODNs induce high IFN-α production from pDC, yet are weak stimulators of TLR9-dependent nuclear factor-κB signaling and B-cell activation. In contrast, the B class strongly activate B cells, but stimulate only low levels of IFN-α secretion. C class CpG ODNs show a combination of the characteristics of the A and B classes, stimulating strong IFN-α production and B-cell stimulation. The immune stimulatory effects of all three ODN classes require TLR9. There is some evidence that their different immune effects may be related to different intracellular compartmentalization of the ODN classes (12). Surprisingly, phosphorothioate ODNs with no CpG motif also stimulate cells through TLR9, but induce little or no IFN-α secretion, and the net effects in vitro and in vivo are more Th2-like (13). A-class ODN have not progressed into human clinical development, due to their complex structures (4). B-class ODNs are widely used as vaccine adjuvants, and C-class ODNs also have shown activity as vaccine adjuvants in animal models (14, 15), and one is in clinical development as a monotherapy against hepatitis C virus (HCV).

TLR9 triggering with B- or C-class ODNs particularly induces the release of Th1 and Th1-like cytokines and chemokines (4). All type-I IFN subtypes, including IFN-β and IFN-ω and the recently described type III IFN, IL-28A/B, and IL-29, are produced upon CpG stimulation. High levels of IFN-inducible proteins, such as OAS (2′-5′ oligoadenylate synthetase), Mx1 (myxoprotein 1), monocyte chemotactic protein-1, and IFN-γ–inducible protein-10, are secreted. Additional antiinfective cytokines induced by CpG-dependent TLR9 stimulation include IL-18 and tumor necrosis factor (TNF)–related apoptosis-inducing ligand. In mice, TLR9 activation results in high levels of IFN-γ and IL-12 secretion, but in humans this response is much weaker. TLR9 activation of human immune cells induces relatively little secretion of proinflammatory cytokines, such as TNF-α and IL-6, but it directly and indirectly induces immune cells, including B cells, monocytes, pDCs, mDCs, NK, NKT, and T cells, to express multiple activation markers, cytokine and chemokine receptors, costimulatory and major histocompatibility complex molecules, including CD69, CD80, CD86, and class I and II major histocompatibility complex.

THERAPEUTIC APPLICATION OF TLR9 ACTIVATION FOR INFECTIOUS DISEASE MONOTHERAPY

Because the biologic function of TLR9 appears to be to stimulate protective immunity in response to infection by intracellular pathogens, prophylactic or therapeutic treatment with a CpG ODN might protect against an intracellular infectious challenge and/or eliminate a chronic infection. Indeed, studies in mice have demonstrated that the innate immune defenses activated by B-class CpG ODNs (almost no studies have been reported with A or C class), given by injection, inhalation, or even by oral administration, can protect against a wide range of viral, bacterial, and even some parasitic pathogens, including lethal challenge with category A agents or surrogates, such a Bacillus anthracis, vaccinia virus, Francisella tularensis, and ebola, as well as more common pathogens, such as Listeria monocytogenes, Mycobacterium tuberculosis, and influenza virus (1633) (Table 1). The mechanisms of protection have only been partially investigated. Protection in an L. monocytogenes model has been linked to CpG-activated DCs, which protect naive mice upon adoptive transfer (3436). Additional cell types may also be able to provide some protection, because naive mice that received CpG-pretreated spleen cells depleted of CD11c+ DCs still had a partial survival benefit. In a herpes simplex virus challenge model, mice depleted of pDCs no longer were protected by CpG pretreatment, and IFN-α was also needed, as mice genetically deficient in the type-I IFN receptor were no longer fully protected (37). On the other hand, in an L. monocytogenes challenge model, type I IFN was not required for CpG-induced protection, even though the protection was abolished when pDCs were depleted (38). In this and many other animal models, IFN-γ was found to be critically required for the CpG-induced protection.

TABLE 1.

PATHOGEN CHALLENGE MODELS IN WHICH CpG OLIGODEOXYNUCLEOTIDES HAVE SHOWN PROTECTION

Species Pathogen Class Challenge/Route Reference Numbers
Mouse Bacteria Listeria monocytogenes 16, 19, 73
Francisella tularensis 16
Mycobacterium avium 74
Mycobacterium tuberculosis 31
Helicobacter pylori 75
Burkholderia mallei 76
Polymicrobial sepsis (peritonitis) 26
Bacillus anthracis Lovchik and colleagues (unpublished data)
Parasite/other Leishmania major 18, 29
Plasmodium yoelii 17
Toxoplasma gondii 77
Cryptococcus neoformans 78, 79
Cryptosporidium parvum 80
Scrapie prion 81
Virus Respiratory syncytial virus 30
Tacaribe arenavirus 82
Herpes simplex virus 27
Influenza 83, 84
Orthopoxvirus 22
Friend retrovirus 32
Foot and mouth disease virus 85
Chicken Bacteria Escherichia coli 86, 87
Sheep Virus Bovine herpesvirus-1 88
Monkey Parasite Leishmania major 89, 90

Postexposure therapy with TLR9 activation is generally ineffective against rapidly progressive acute infectious agents. However, there may be a role for TLR9 activation in the therapy of chronic viral infections, because hepatitis B virus (HBV)–transgenic mice treated with a CpG ODN showed a significant decrease in viral expression (39). As hepatocytes normally do not express TLR9, the antiviral effect in this model is presumably indirect. HBV expression was not suppressed in mice genetically deficient in the type I IFN receptor, suggesting that the antiviral effect of CpG therapy in this model results from the CpG-induced IFN-α secretion, presumably by pDCs.

HCV is an important human pathogen, which chronically infects approximately 170 million people worldwide. Infection can lead to liver cirrhosis and death, and is currently the major cause of liver failure requiring transplantation in North America. Fewer than half of North American patients respond to the current standard-of-care treatment for HCV, which consists of 48 weeks of a combination therapy with IFN-α and ribavirin. In up to 20% of acutely infected patients with HCV, the immune system is able to clear the infection without specific therapy. This spontaneous viral clearance is associated with early and strong innate immune activation, leading to the development of a strong and diverse adaptive immune response with anti-HCV Th1 and CD8 cytolytic T cells (40). Because TLR9 activation can drive a similar pattern of innate and adaptive immune responses to that seen in the spontaneous resolvers, we investigated whether a C-class CpG ODN, CPG 10101, may have activity against HCV. In a 4-week, phase Ib, blinded, randomized, controlled trial involving 60 HCV-infected subjects, monotherapy with once- or twice-weekly subcutaneous injection of CPG 10101 caused a dose-dependent decrease in blood viral RNA levels (41). At the highest dose level of 0.75 mg/kg weekly, there was a 1.6 mean log reduction in viral RNA, which was associated with biomarkers for TLR9 activation, including NK cell activation and serum IFN-α and IFN-inducible chemokines. Treatment was generally well tolerated, with the most common side effects being mild to moderate flulike symptoms and injection site reactions, and the maximal tolerated dose was not reached. This trial showed encouraging anti-HCV activity for CpG 10101 as a monotherapy. Based on in vitro studies showing that exogenous IFN-α primes human peripheral blood mononuclear cells (PBMCs) for stronger responses to CpG (42), and other studies suggesting synergy between TLR9 activation and ribavirin, we decided to perform a clinical trial using the CPG 10101 in a combination regimen, together with the conventional, partially effective therapy of pegylated IFN and ribavirin. The randomized phase Ib clinical study enrolled 74 evaluable genotype-1 patients chronically infected with HCV. Subjects had all previously received at least 24 weeks of treatment with the standard of care (pegylated IFN and ribavirin), and achieved viral negativity, but had subsequently relapsed within 6 months of treatment. Patients in the study were randomly assigned to 1 of 5 groups, receiving 12 weekly doses of CPG 10101 alone, CPG 10101 in combination with pegylated IFN, CPG 10101 with ribavirin, CPG 10101 with pegylated IFN and ribavirin, or pegylated IFN and ribavirin. CPG 10101 was administered by subcutaneous injection at a dose of 0.2 mg/kg once weekly. At 12 weeks, 50% (7/14) of treatment-refractory patients in the CPG 10101–pegylated IFN–ribavirin arm of the study achieved undetectable HCV RNA levels, or viral negativity versus only 2 of 13 of those patients who received pegylated IFN and ribavirin alone (p = 0.050). Unfortunately, nearly all of these subjects relapsed after discontinuation of treatment. The triplet combination of CPG 10101 with pegylated IFN and ribavirin resulted in a 3.3 mean log reduction in HCV RNA levels versus a 2.3 mean log reduction (p < 0.050) among patients receiving the control combination. The CPG 10101 combinations were generally well tolerated. Adverse events were similar to pegylated IFN and ribavirin treatment, were predominantly mild to moderate in intensity, and consisted of flulike symptoms, headache, and injection-site reactions.

APPLICATIONS OF TLR9 ACTIVATION FOR ENHANCING INFECTIOUS DISEASE VACCINES

CpG ODNs have become well established as a gold standard vaccine adjuvant, capable of inducing powerful antigen-specific antibody and Th1 cellular immune responses in many vertebrate species, including humans. The range of vaccines in which this has been demonstrated include peptide or protein antigens, live or killed viruses, DC vaccines, autologous cellular vaccines, and polysaccharide conjugates. The strong vaccine adjuvant activity of CpG ODN presumably results from some or all of the following factors: (1) synergy between TLR9 and B-cell receptor preferentially stimulates antigen-specific B cells (43); (2) inhibition of B-cell apoptosis (44); (3) enhanced IgG class switch DNA recombination (4547); and (4) DC maturation and differentiation, resulting in enhanced activation of Th1 cells and strong cytotoxic T lymphocyte (CTL) generation, even in the absence of CD4 T-cell help (48, 49). CpG ODNs show even greater adjuvant activity when formulated or coadministered with other adjuvants or in formulations such as microparticles, nanoparticles, lipid emulsions, or similar formulations (50). In humans, CpG ODNs have been used as adjuvants for hepatitis B vaccination, either in combination with alum (51) or alone (52). In a randomized, double-blind, controlled, phase I/II dose escalation study, healthy individuals received three intramuscular injections (using the U.S. Food and Drug Administration–approved vaccination regimen of 0, 4, and 24 wk) of an alum-absorbed HBV vaccine, either in saline or mixed with a B-class ODN, CPG 7909, at doses of 0.125, 0.5, or 1.0 mg (51). HBsAg-specific antibody responses (anti-HBs) appeared earlier and had higher titers at all time points from 2 weeks after the initial prime up to 48 weeks in CPG 7909 recipients compared to those individuals who received vaccine alone. Moreover, most of the subjects who received CPG 7909 as adjuvant developed protective levels of anti-HBs IgG within just 2 weeks of the priming vaccine dose, compared to none of the subjects receiving the commercial vaccine alone (51). The addition of the CpG ODNs also improved the quality of the antigen-specific antibody response, with an increased proportion of high-avidity antibodies (53). The ability of CPG 7909 to accelerate seroconversion has also been demonstrated when used as an adjuvant to the approved anthrax vaccine in a randomized, controlled trial in healthy volunteers. Control subjects reached their peak titer of toxin-neutralizing antibody at Day 46, but this titer was achieved in the subjects receiving CPG 7909 already at Day 22, more than 3 weeks earlier (54). More rapid seroconversion to the anthrax toxin could be of great importance in the setting of a bioterror attack. Furthermore, the addition of CPG 7909 induced a statistically significant, 8.8-fold increase in the peak titer of toxin-neutralizing antibody, and increased the proportion of subjects who achieved a strong IgG response to the anthrax-protective antigen from 61 to 100% (54). These results indicate great potential for TLR9 agonists as vaccine adjuvants.

Certain populations are hyporesponsive to vaccination, especially immune-suppressed individuals, such as those infected with HIV. A randomized, double-blind, controlled trial in HIV-infected humans demonstrated that addition of CPG 7909 to the Engerix B vaccine significantly enhanced both the mean titers of anti-HBs and the antigen-specific T-cell proliferative response (55). The proportion of patients with HIV who had seroprotective levels at 12 months after vaccination was increased from 63% in the control subjects to 100% in the group receiving CPG 7909 (55).

One of the limitations in vaccine development is the cost of antigen production, especially for vaccines such as the flu vaccine, which have to be produced in large quantities in a short time frame. The use of a CpG ODN as vaccine adjuvant in mice enables the antigen doses to be reduced by approximately two orders of magnitude, with comparable antibody responses to the full-dose vaccine without CpG (56). In a phase Ib randomized, double-blind, controlled clinical trial, subjects vaccinated with a one-tenth dose of a commercial, trivalent, killed, split influenza vaccine (Fluarix) had reduced levels of antigen-specific IFN-γ secretion from restimulated PBMCs compared with those measured in PBMCs from subjects vaccinated with the full-dose vaccine alone (57). However, the coadministration of CPG 7909 with the one-tenth dose of Fluarix restored the antigen-specific IFN-γ secretion to the level seen with full-dose vaccine (57). This suggests that addition of a CpG ODN to a flu vaccine could enable the effective use of the vaccine with lower antigen doses.

SAFETY OF CpG ODNs

Even if they do not contain a CpG motif, all phosphorothioate backbone (PS) ODNs can have a variety of sequence-independent, backbone-related effects that have been characterized in detailed studies of antisense ODNs (5860). These effects are most prominent in rodents, which show, on chronic dosing of ODNs, dose-dependent mononuclear cell infiltration in the organs of ODN deposition (58, 61), and largely depend on TLR9 (58, 62). Such changes have not been described in monkeys or humans. Presumably these species-specific findings are a consequence of the cellular pattern of TLR9 expression, which determines the cytokines that will be produced in response to administration of a CpG ODN, and thus, the safety profile of the drug. Because TLR9 is expressed in a broader range of immune cells in rodents compared with primates, the rodent tends to overpredict toxicities that will occur in primates. For example, rodents respond to CpG ODN administration with high serum concentrations of proinflammatory cytokines, such as TNF-α, which can result in a lethal “cytokine storm” (63), but in humans and primates there is no change in serum TNF-α after CpG injection, which is generally well tolerated (64).

In terms of mechanism of action-related effects, CpG ODN treatment clearly can exacerbate autoimmunity in mouse models of lupus (65), multiple sclerosis (66), colitis (67), and arthritis (68). On the other hand, CpG ODNs protect against autoimmunity and inflammatory diseases in other murine experimental systems through mechanisms ranging from induction of IFN-γ secretion to expression of IDO (6972). Regardless of the effects in those models, there has, to date, been no case of human systemic autoimmune disease attributed to treatment with a CpG ODN.

The safety profile of several TLR9 agonists in man has been observed in the clinical trials described above over a more than 1,000-fold dose range, from 0.0025 to 0.81 mg/kg. A maximal tolerated dose in humans has not been reported to date. The primary adverse events are dose-dependent local injection reactions (e.g., erythema, pain, swelling, induration, pruritus, or warmth at the site of injection) or systemic, flulike reactions (e.g., headache, rigors, myalgia, pyrexia, nausea, and vomiting), and are consistent with the known TLR9 agonist mechanism of action. Depending on the dose, systemic symptoms typically appear within 12 to 24 hours of dosing and persist for 1 to 2 days. At the low doses used in vaccine trials, there appears to be a slight increase in the frequency of injection site reactions, which are generally mild, above the frequency observed with the vaccine alone. So far, no subjects have been reported to have developed an autoimmune disease after CpG therapy, but the duration of therapy has usually been less than 6 months; only a few patients have received chronic therapy with CpG ODNs for longer than 3 years. Definite conclusions on the safety of chronic TLR9 activation with CpG ODNs await the completion of clinical trials involving larger numbers of patients followed for longer periods of time.

CONCLUSIONS

TLR9 activation with synthetic CpG ODNs induces powerful Th1-like innate and adaptive immune responses, and is emerging as a promising therapeutic approach in several disease fields. Treatment with CpG ODNs can provide a temporary protection against diverse pathogen challenges in mice, chickens, sheep, and nonhuman primates, and reduces HCV RNA levels in chronically infected humans. CpG ODNs have become recognized as powerful vaccine adjuvants, inducing faster and stronger humoral and cellular immune responses. To date, the safety of TLR9 activation with CpG ODNs appears good, but larger clinical trials and a longer duration of follow-up will be needed to determine the place of these new potential agents in the therapeutic armamentarium.

Acknowledgments

The author thanks Meredith Larade for expert assistance with manuscript preparation.

Supported by National Institute of Allergy and Infectious Diseases grant U01 AI057264 and the Defense Advanced Research Projects Agency.

Conflict of Interest Statement: A.M.K. is employed by Coley Pharmaceutical Group and is an inventor on patents relating to this technology, as well as a shareholder in Coley, which is seeking to commercialize the technology.

References

  • 1.Pulendran B. Variegation of the immune response with dendritic cells and pathogen recognition receptors. J Immunol 2005;174:2457–2465. [DOI] [PubMed] [Google Scholar]
  • 2.Ishii KJ, Akira S. Innate immune recognition of, and regulation by, DNA. Trends Immunol 2006;27:525–532. [DOI] [PubMed] [Google Scholar]
  • 3.Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, Matsumoto M, Hoshino K, Wagner H, Takeda K, et al. A Toll-like receptor recognizes bacterial DNA. Nature 2000;408:740–745. (In Process Citation). [DOI] [PubMed] [Google Scholar]
  • 4.Krieg AM. Therapeutic potential of Toll-like receptor 9 activation. Nat Rev Drug Discov 2006;5:471–484. [DOI] [PubMed] [Google Scholar]
  • 5.Liu YJ. IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu Rev Immunol 2005;23:275–306. [DOI] [PubMed] [Google Scholar]
  • 6.Krieg AM. CpG motifs in bacterial DNA and their immune effects. Annu Rev Immunol 2002;20:709–760. [DOI] [PubMed] [Google Scholar]
  • 7.Jung J, Yi AK, Zhang X, Choe J, Li L, Choi YS. Distinct response of human B cell subpopulations in recognition of an innate immune signal, CpG DNA. J Immunol 2002;169:2368–2373. [DOI] [PubMed] [Google Scholar]
  • 8.Bernasconi NL, Traggiai E, Lanzavecchia A. Maintenance of serological memory by polyclonal activation of human memory B cells. Science 2002;298:2199–2202. [DOI] [PubMed] [Google Scholar]
  • 9.Bernasconi NL, Onai N, Lanzavecchia A. A role for Toll-like receptors in acquired immunity: up-regulation of TLR9 by BCR triggering in naive B cells and constitutive expression in memory B cells. Blood 2003;101:4500–4504. [DOI] [PubMed] [Google Scholar]
  • 10.Traggiai E, Becker S, Subbarao K, Kolesnikova L, Uematsu Y, Gismondo MR, Murphy BR, Rappuoli R, Lanzavecchia A. An efficient method to make human monoclonal antibodies from memory B cells: potent neutralization of SARS coronavirus. Nat Med 2004;10:871–875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Poeck H, Wagner M, Battiany J, Rothenfusser S, Wellisch D, Hornung V, Jahrsdorfer B, Giese T, Endres S, Hartmann G. Plasmacytoid dendritic cells, antigen and CpG-C license human B cells for plasma cell differentiation and immunoglobulin production in the absence of T cell help. Blood 2004;103:3058–3064. [DOI] [PubMed] [Google Scholar]
  • 12.Honda K, Ohba Y, Yanai H, Negishi H, Mizutani T, Takaoka A, Taya C, Taniguchi T. Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction. Nature 2005;434:1035–1040. [DOI] [PubMed] [Google Scholar]
  • 13.Vollmer J, Weeratna R, Payette P, Jurk M, Schetter C, Laucht M, Wader T, Tluk S, Liu M, Davis HL, et al. Characterization of three CpG oligodeoxynucleotide classes with distinct immunostimulatory activities. Eur J Immunol 2004;34:251–262. [DOI] [PubMed] [Google Scholar]
  • 14.Gu M, Hine PM, James JW, Giri L, Nabors GS. Increased potency of BioThrax anthrax vaccine with the addition of the C-class CpG oligonucleotide adjuvant CPG 10109. Vaccine 2007;25:526–534. [DOI] [PubMed] [Google Scholar]
  • 15.Wille-Reece U, Flynn BJ, Lore K, Koup RA, Miles AP, Saul A, Kedl RM, Mattapallil JJ, Weiss WR, Roederer M, et al. Toll-like receptor agonists influence the magnitude and quality of memory T cell responses after prime-boost immunization in nonhuman primates. J Exp Med 2006;203:1249–1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Elkins KL, Rhinehart-Jones TR, Stibitz S, Conover JS, Klinman DM. Bacterial DNA containing CpG motifs stimulates lymphocyte-dependent protection of mice against lethal infection with intracellular bacteria. J Immunol 1999;162:2291–2298. [PubMed] [Google Scholar]
  • 17.Gramzinski RA, Doolan DL, Sedegah M, Davis HL, Krieg AM, Hoffman SL. Interleukin-12– and γ interferon–dependent protection against malaria conferred by CpG oligodeoxynucleotide in mice. Infect Immun 2001;69:1643–1649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zimmermann S, Egeter O, Hausmann S, Lipford GB, Rocken M, Wagner H, Heeg K. CpG oligodeoxynucleotides trigger protective and curative Th1 responses in lethal murine Leishmaniasis. J Immunol 1998;160:3627–3630. [PubMed] [Google Scholar]
  • 19.Krieg AM, Love-Homan L, Yi AK, Harty JT. CpG DNA induces sustained IL-12 expression in vivo and resistance to Listeria monocytogenes challenge. J Immunol 1998;161:2428–2434. [PubMed] [Google Scholar]
  • 20.Klinman DM, Conover J, Coban C. Repeated administration of synthetic oligodeoxynucleotides expressing CpG motifs provides long-term protection against bacterial infection. Infect Immun 1999;67:5658–5663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Klinman DM, Verthelyi D, Takeshita F, Ishii KJ. Immune recognition of foreign DNA: a cure for bioterrorism? Immunity 1999;11:123–129. [DOI] [PubMed] [Google Scholar]
  • 22.Rees DG, Gates AJ, Green M, Eastaugh L, Lukaszewski RA, Griffin KF, Krieg AM, Titball RW. CpG-DNA protects against a lethal orthopoxvirus infection in a murine model. Antiviral Res 2005;65:87–95. [DOI] [PubMed] [Google Scholar]
  • 23.Deng JC, Moore TA, Newstead MW, Zeng X, Krieg AM, Standiford TJ. CpG oligodeoxynucleotides stimulate protective innate immunity against pulmonary Klebsiella infection. J Immunol 2004;173:5148–5155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ray NB, Krieg AM. Oral pretreatment of mice with CpG DNA reduces susceptibility to oral or intraperitoneal challenge with virulent Listeria monocytogenes. Infect Immun 2003;71:4398–4404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Klinman DM. Immunotherapeutic uses of CpG oligodeoxynucleotides. Nat Rev Immunol 2004;4:249–259. [DOI] [PubMed] [Google Scholar]
  • 26.Weighardt H, Feterowski C, Veit M, Rump M, Wagner H, Holzmann B. Increased resistance against acute polymicrobial sepsis in mice challenged with immunostimulatory CpG oligodeoxynucleotides is related to an enhanced innate effector cell response. J Immunol 2000;165:4537–4543. [DOI] [PubMed] [Google Scholar]
  • 27.Pyles RB, Higgins D, Chalk C, Zalar A, Eiden J, Brown C, Van Nest G, Stanberry LR. Use of immunostimulatory sequence–containing oligonucleotides as topical therapy for genital herpes simplex virus type 2 infection. J Virol 2002;76:11387–11396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ashkar AA, Bauer S, Mitchell WJ, Vieira J, Rosenthal KL. Local delivery of CpG oligodeoxynucleotides induces rapid changes in the genital mucosa and inhibits replication, but not entry, of herpes simplex virus type 2. J Virol 2003;77:8948–8956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Walker PS, Scharton-Kersten T, Krieg AM, Love-Homan L, Rowton ED, Udey MC, Vogel JC. Immunostimulatory oligodeoxynucleotides promote protective immunity and provide systemic therapy for Leishmaniasis via IL-12– and IFN-γ–dependent mechanisms. Proc Natl Acad Sci USA 1999;96:6970–6975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Cho JY, Miller M, Baek KJ, Castaneda D, Nayar J, Roman M, Raz E, Broide DH. Immunostimulatory DNA sequences inhibit respiratory syncytial viral load, airway inflammation, and mucus secretion. J Allergy Clin Immunol 2001;108:697–702. [DOI] [PubMed] [Google Scholar]
  • 31.Juffermans NP, Leemans JC, Florquin S, Verbon A, Kolk AH, Speelman P, van Deventer SJ, van der Poll T. CpG oligodeoxynucleotides enhance host defense during murine tuberculosis. Infect Immun 2002;70:147–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Olbrich AR, Schimmer S, Heeg K, Schepers K, Schumacher TN, Dittmer U. Effective postexposure treatment of retrovirus-induced disease with immunostimulatory DNA containing CpG motifs. J Virol 2002;76:11397–11404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Freidag BL, Melton GB, Collins F, Klinman DM, Cheever A, Stobie L, Suen W, Seder RA. CpG oligodeoxynucleotides and interleukin-12 improve the efficacy of Mycobacterium bovis BCG vaccination in mice challenged with M. tuberculosis. Infect Immun 2000;68:2948–2953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ramirez-Pineda JR, Frohlich A, Berberich C, Moll H. Dendritic cells (DC) activated by CpG DNA ex vivo are potent inducers of host resistance to an intracellular pathogen that is independent of IL-12 derived from the immunizing DC. J Immunol 2004;172:6281–6289. [DOI] [PubMed] [Google Scholar]
  • 35.Ishii KJ, Ito S, Tamura T, Hemmi H, Conover J, Ozato K, Akira S, Klinman DM. CpG-activated Thy1.2+ dendritic cells protect against lethal Listeria monocytogenes infection. Eur J Immunol 2005;35:2397–2405. [DOI] [PubMed] [Google Scholar]
  • 36.Lugo-Villarino G, Ito S, Klinman DM, Glimcher LH. The adjuvant activity of CpG DNA requires T-bet expression in dendritic cells. Proc Natl Acad Sci USA 2005;102:13248–13253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Shen H, Iwasaki A. A crucial role for plasmacytoid dendritic cells in antiviral protection by CpG ODN–based vaginal microbicide. J Clin Invest 2006;116:2237–2243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kuwajima S, Sato T, Ishida K, Tada H, Tezuka H, Ohteki T. Interleukin 15–dependent crosstalk between conventional and plasmacytoid dendritic cells is essential for CpG-induced immune activation. Nat Immunol 2006;7:740–746. [DOI] [PubMed] [Google Scholar]
  • 39.Isogawa M, Robek MD, Furuichi Y, Chisari FV. Toll-like receptor signaling inhibits hepatitis B virus replication in vivo. J Virol 2005;79:7269–7272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rehermann B, Nascimbeni M. Immunology of hepatitis B virus and hepatitis C virus infection. Nat Rev Immunol 2005;5:215–229. [DOI] [PubMed] [Google Scholar]
  • 41.McHutchison JG, Bacon BR, Gordon SC, Lawitz E, Shiffman M, Afdhal NH, Jacobson IM, Muir A, Efler SM, Al-Adhami M, et al. Relationships of HCV RNA responses to CPG 10101, a TLR9 agonist: pharmacodynamics & patient characteristics. Hepatology 2005;42:249A.16025494 [Google Scholar]
  • 42.Vallin H, Perers A, Alm GV, Ronnblom L. Anti–double-stranded DNA antibodies and immunostimulatory plasmid DNA in combination mimic the endogenous IFN-α inducer in systemic lupus erythematosus. J Immunol 1999;163:6306–6313. [PubMed] [Google Scholar]
  • 43.Krieg AM, Yi AK, Matson S, Waldschmidt TJ, Bishop GA, Teasdale R, Koretzky GA, Klinman DM. CpG motifs in bacterial DNA trigger direct B-cell activation. Nature 1995;374:546–549. [DOI] [PubMed] [Google Scholar]
  • 44.Yi AK, Chang M, Peckham DW, Krieg AM, Ashman RF. CpG oligodeoxyribonucleotides rescue mature spleen B cells from spontaneous apoptosis and promote cell cycle entry. J Immunol 1998;160:5898–5906. [published erratum appears in J Immunol 163:1093.] [PubMed] [Google Scholar]
  • 45.Davis HL, Weeratna R, Waldschmidt TJ, Tygrett L, Schorr J, Krieg AM, Weeratna R. CpG DNA is a potent enhancer of specific immunity in mice immunized with recombinant hepatitis B surface antigen. J Immunol 1998;160:870–876. [published erratum appears in J Immunol 162:3103.] [PubMed] [Google Scholar]
  • 46.Liu N, Ohnishi N, Ni L, Akira S, Bacon KB. CpG directly induces T-bet expression and inhibits IgG1 and IgE switching in B cells. Nat Immunol 2003;4:687–693. [DOI] [PubMed] [Google Scholar]
  • 47.He B, Qiao X, Cerutti A. CpG DNA induces IgG class switch DNA recombination by activating human B cells through an innate pathway that requires TLR9 and cooperates with IL-10. J Immunol 2004;173:4479–4491. [DOI] [PubMed] [Google Scholar]
  • 48.Lipford GB, Sparwasser T, Zimmermann S, Heeg K, Wagner H. CpG-DNA–mediated transient lymphadenopathy is associated with a state of Th1 predisposition to antigen-driven responses. J Immunol 2000;165:1228–1235. [DOI] [PubMed] [Google Scholar]
  • 49.Sparwasser T, Vabulas RM, Villmow B, Lipford GB, Wagner H. Bacterial CpG-DNA activates dendritic cells in vivo: T helper cell–independent cytotoxic T cell responses to soluble proteins. Eur J Immunol 2000;30:3591–3597. [DOI] [PubMed] [Google Scholar]
  • 50.Krieg AM, Davis HL. Enhancing vaccines with immune stimulatory CpG DNA. Curr Opin Mol Ther 2001;3:15–24. [PubMed] [Google Scholar]
  • 51.Cooper CL, Davis H, Morris ML, Efler SM, Al Adhami MJ, Krieg A, Cameron DW, Heathcoat J. CpG 7909, an immunostimulatory TLR9 agonist oligodeoxynucleotide, as adjuvant to Engerix-B HBV vaccine in healthy adults: a double-blind phase I/II study. J Clin Immunol 2004;24:693–702. [DOI] [PubMed] [Google Scholar]
  • 52.Halperin SA, Van Nest G, Smith B, Abtahi S, Whiley H, Eiden JJ. A phase I study of the safety and immunogenicity of recombinant hepatitis B surface antigen co-administered with an immunostimulatory phosphorothioate oligonucleotide adjuvant. Vaccine 2003;21:2461–2467. [DOI] [PubMed] [Google Scholar]
  • 53.Siegrist CA, Pihlgren M, Tougne C, Efler SM, Morris ML, Al Adhami MJ, Cameron DW, Cooper CL, Heathcote J, Davis HL, et al. Co-administration of CpG oligonucleotides enhances the late affinity maturation process of human anti–hepatitis B vaccine response. Vaccine 2004;23:615–622. [DOI] [PubMed] [Google Scholar]
  • 54.Rynkiewicz D, Rathkopf M, Ransom J, Sim I, Giri L, Quinn J, Waytes T, Al-Adhami M, Johnson W, Nielsen C. Marked enhancement of antibody response to anthrax vaccine adsorbed with CPG 7909 in healthy volunteers. [LB25] ICAAC 2005. Abstracts and Final Program of the Interscience Conference on Antimicrobial Agents and Chemotherapy; 2005 Dec 16–19; Washington, DC. Herndon (VA): ASM Press.
  • 55.Cooper CL, Davis HL, Angel JB, Morris ML, Elfer SM, Seguin I, Krieg AM, Cameron DW. CPG 7909 adjuvant improves hepatitis B virus vaccine seroprotection in antiretroviral-treated HIV-infected adults. AIDS 2005;19:1473–1479. [DOI] [PubMed] [Google Scholar]
  • 56.Weeratna R, Comanita L, Davis HL. CPG ODN allows lower dose of antigen against hepatitis B surface antigen in BALB/c mice. Immunol Cell Biol 2003;81:59–62. [DOI] [PubMed] [Google Scholar]
  • 57.Cooper CL, Davis H, Morris ML, Efler SM, Krieg A, Li Y, Laframboise C, Al Adhami MJ, Khaliq Y, Sequin I, et al. Safety and immunogenicity of CpG 7909 injection as an adjuvant to fluarix influenza vaccine. Vaccine 2004;22:3136–3143. [DOI] [PubMed] [Google Scholar]
  • 58.Levin AA, Henry S, Monteith D. Toxicity of antisense oligonucleotides. In: Crooke ST, editor. Antisense drug technology. New York: Marcel Dekker; 2001. pp. 201–267.
  • 59.Levin AA. A review of the issues in the pharmacokinetics and toxicology of phosphorothioate antisense oligonucleotides. Biochim Biophys Acta 1999;1489:69–84. [DOI] [PubMed] [Google Scholar]
  • 60.Monteith DK, Levin AA. Synthetic oligonucleotides: the development of antisense therapeutics. Toxicol Pathol 1999;27:8–13. [DOI] [PubMed] [Google Scholar]
  • 61.Henry SP, Taylor J, Midgley L, Levin AA, Kornbrust DJ. Evaluation of the toxicity of ISIS 2302, a phosphorothioate oligonucleotide, in a 4-week study in CD-1 mice. Antisense Nucleic Acid Drug Dev 1997;7:473–481. [DOI] [PubMed]
  • 62.Vollmer J, Jurk M, Samulowitz U, Lipford G, Forsbach A, Wullner M, Tluk S, Hartmann H, Kritzler A, Muller C, et al. CpG oligodeoxynucleotides stimulate IFN-gamma-inducible protein-10 production in human B cells. J Endotoxin Res 2004;10:431–438. [DOI] [PubMed] [Google Scholar]
  • 63.Sparwasser T, Miethke T, Lipford G, Borschert K, Hacker H, Heeg K, Wagner H. Bacterial DNA causes septic shock [letter]. Nature 1997;386:336–337. [DOI] [PubMed] [Google Scholar]
  • 64.Krieg AM, Efler SM, Wittpoth M, Al Adhami MJ, Davis HL. Induction of systemic Th1-like innate immunity in normal volunteers following subcutaneous but not intravenous administration of CPG 7909, a synthetic B-class CpG oligodeoxynucleotide TLR9 agonist. J Immunother 2004;27:460–471. [DOI] [PubMed] [Google Scholar]
  • 65.Hasegawa K, Hayashi T. Synthetic CpG oligodeoxynucleotides accelerate the development of lupus nephritis during preactive phase in NZB x NZWF1 mice. Lupus 2003;12:838–845. [DOI] [PubMed] [Google Scholar]
  • 66.Ichikawa HT, Williams LP, Segal BM. Activation of APCs through CD40 or Toll-like receptor 9 overcomes tolerance and precipitates autoimmune disease. J Immunol 2002;169:2781–2787. [DOI] [PubMed] [Google Scholar]
  • 67.Obermeier F, Dunger N, Deml L, Herfarth H, Scholmerich J, Falk W. CpG motifs of bacterial DNA exacerbate colitis of dextran sulfate sodium–treated mice. Eur J Immunol 2002;32:2084–2092. [DOI] [PubMed] [Google Scholar]
  • 68.Ronaghy A, Prakken BJ, Takabayashi K, Firestein GS, Boyle D, Zvailfler NJ, Roord ST, Albani S, Carson DA, Raz E. Immunostimulatory DNA sequences influence the course of adjuvant arthritis. J Immunol 2002;168:51–56. [DOI] [PubMed] [Google Scholar]
  • 69.Boccaccio GL, Mor F, Steinman L. Non-coding plasmid DNA induces IFN-γ in vivo and suppresses autoimmune encephalomyelitis. Int Immunol 1999;11:289–296. [DOI] [PubMed] [Google Scholar]
  • 70.Quintana FJ, Rotem A, Carmi P, Cohen IR. Vaccination with empty plasmid DNA or CpG oligonucleotide inhibits diabetes in nonobese diabetic mice: modulation of spontaneous 60-kDa heat shock protein autoimmunity. J Immunol 2000;165:6148–6155. [DOI] [PubMed] [Google Scholar]
  • 71.Hayashi T, Beck L, Rossetto C, Gong X, Takikawa O, Takabayashi K, Broide DH, Carson DA, Raz E. Inhibition of experimental asthma by indoleamine 2,3-dioxygenase. J Clin Invest 2004;114:270–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Katakura K, Lee J, Rachmilewitz D, Li G, Eckmann L, Raz E. Toll-like receptor 9–induced type I IFN protects mice from experimental colitis. J Clin Invest 2005;115:695–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Oxenius A, Martinic MM, Hengartner H, Klenerman P. CpG-containing oligonucleotides are efficient adjuvants for induction of protective antiviral immune responses with T-cell peptide vaccines. J Virol 1999;73:4120–4126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hayashi T, Rao SP, Takabayashi K, Van Uden JH, Kornbluth RS, Baird SM, Taylor MW, Carson DA, Catanzaro A, Raz E. Enhancement of innate immunity against Mycobacterium avium infection by immunostimulatory DNA is mediated by indoleamine 2,3-dioxygenase. Infect Immun 2001;69:6156–6164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Raghavan S, Nystrom J, Fredriksson M, Holmgren J, Harandi AM. Orally administered CpG oligodeoxynucleotide induces production of CXC and CC chemokines in the gastric mucosa and suppresses bacterial colonization in a mouse model of Helicobacter pylori infection. Infect Immun 2003;71:7014–7022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Waag DM, McCluskie MJ, Zhang N, Krieg AM. A CpG oligonucleotide can protect mice from a low aerosol challenge dose of Burkholderia mallei. Infect Immun 2006;74:1944–1948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Saavedra R, Leyva R, Tenorio EP, Haces ML, Rodriguez-Sosa M, Terrazas LI, Herion P. CpG-containing ODN has a limited role in the protection against Toxoplasma gondii. Parasite Immunol 2004;26:67–73. [DOI] [PubMed] [Google Scholar]
  • 78.Miyagi K, Kawakami K, Kinjo Y, Uezu K, Kinjo T, Nakamura K, Saito A. CpG oligodeoxynucleotides promote the host protective response against infection with Cryptococcus neoformans through induction of interferon-γ production by CD4+ T cells. Clin Exp Immunol 2005;140:220–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Edwards L, Williams AE, Krieg AM, Rae AJ, Snelgrove RJ, Hussell T. Stimulation via Toll-like receptor 9 reduces Cryptococcus neoformans–induced pulmonary inflammation in an IL-12–dependent manner. Eur J Immunol 2005;35:273–281. [DOI] [PubMed] [Google Scholar]
  • 80.Barrier M, Lacroix-Lamande S, Mancassola R, Auray G, Bernardet N, Chausse AM, Uematsu S, Akira S, Laurent F. Oral and intraperitoneal administration of phosphorothioate oligodeoxynucleotides leads to control of Cryptosporidium parvum infection in neonatal mice. J Infect Dis 2006;193:1400–1407. [DOI] [PubMed] [Google Scholar]
  • 81.Sethi S, Lipford G, Wagner H, Kretzschmar H. Postexposure prophylaxis against prion disease with a stimulator of innate immunity. Lancet 2002;360:229–230. [DOI] [PubMed] [Google Scholar]
  • 82.Pedras-Vasconcelos JA, Goucher D, Puig M, Tonelli LH, Wang V, Ito S, Verthelyi D. CpG oligodeoxynucleotides protect newborn mice from a lethal challenge with the neurotropic Tacaribe arenavirus. J Immunol 2006;176:4940–4949. [DOI] [PubMed] [Google Scholar]
  • 83.Dong L, Mori I, Hossain MJ, Liu B, Kimura Y. An immunostimulatory oligodeoxynucleotide containing a cytidine-guanosine motif protects senescence-accelerated mice from lethal influenza virus by augmenting the T helper type 1 response. J Gen Virol 2003;84:1623–1628. [DOI] [PubMed] [Google Scholar]
  • 84.Wong JP, Nagata LP, Christopher ME, Salazar AM, Dale RM. Prophylaxis of acute respiratory virus infections using nucleic acid–based drugs. Vaccine 2005;23:2266–2268. [DOI] [PubMed] [Google Scholar]
  • 85.Kamstrup S, Frimann TH, Barfoed AM. Protection of Balb/c mice against infection with FMDV by immunostimulation with CpG oligonucleotides. Antiviral Res 2006;72:42–48. [DOI] [PubMed] [Google Scholar]
  • 86.Gomis S, Babiuk L, Godson DL, Allan B, Thrush T, Townsend H, Willson P, Waters E, Hecker R, Potter A. Protection of chickens against Escherichia coli infections by DNA containing CpG motifs. Infect Immun 2003;71:857–863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Gomis S, Babiuk L, Allan B, Willson P, Waters E, Ambrose N, Hecker R, Potter A. Protection of neonatal chicks against a lethal challenge of Escherichia coli using DNA containing cytosine-phosphodiester-guanine motifs. Avian Dis 2004;48:813–822. [DOI] [PubMed] [Google Scholar]
  • 88.Nichani AK, Mena A, Kaushik RS, Mutwiri GK, Townsend HG, Hecker R, Krieg AM, Babiuk LA, Griebel PJ. Stimulation of innate immune responses by CpG oligodeoxynucleotide in newborn lambs can reduce bovine herpesvirus-1 shedding. Oligonucleotides 2006;16:58–67. [DOI] [PubMed] [Google Scholar]
  • 89.Flynn B, Wang V, Sacks DL, Seder RA, Verthelyi D. Prevention and treatment of cutaneous Leishmaniasis in primates by using synthetic type D/A oligodeoxynucleotides expressing CpG motifs. Infect Immun 2005;73:4948–4954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Verthelyi D, Gursel M, Kenney RT, Lifson JD, Liu S, Mican J, Klinman DM. CpG oligodeoxynucleotides protect normal and SIV-infected macaques from Leishmania infection. J Immunol 2003;170:4717–4723. [DOI] [PubMed] [Google Scholar]

Articles from Proceedings of the American Thoracic Society are provided here courtesy of American Thoracic Society

RESOURCES