651
Views
12
CrossRef citations to date
0
Altmetric
Special Focus Review

Genetic vaccination for re-establishing T-cell tolerance in type 1 diabetes

, &
Pages 27-36 | Received 16 Jun 2010, Accepted 28 Jun 2010, Published online: 01 Jan 2011

Abstract

Type 1 diabetes (T1D) is a T-cell mediated autoimmune disease resulting in the destruction of the insulin-secreting β cells. Currently, there is no established clinical approach to effectively suppress long-term the diabetogenic response. Genetic-based vaccination offers a general strategy to reestablish β-cell specific tolerance within the T-cell compartment. The transfer of genes encoding β-cell autoantigens, anti-inflammatory cytokines and/or immunomodulatory proteins has proven to be effective at preventing and suppressing the diabetogenic response in animal models of T1D. The current review will discuss genetic approaches to prevent and treat T1D with an emphasis on plasmid DNA- and adeno-associated virus-based vaccines.

Introduction

Type 1 diabetes (T1D) is characterized by the autoimmune-mediated destruction of the insulin producing β cells residing in the pancreatic islets of Langerhans.Citation1Citation4 The disease process is viewed as a chronic inflammatory response of the islets, typically progressing over a number of years until the functional mass of β cells is insufficient to meet the body's insulin needs. It is well established from studies carried out in spontaneous rodent models of T1D, such as the nonobese diabetic (NOD) mouse, that the primary mediators of β cell destruction are CD4+ and CD8+ T cells.Citation5Citation7 Indirect evidence for a role for T cells in human T1D is provided by detection of increased β cell-specific CD4+ and CD8+ T cells in peripheral blood lymphocytes of at risk and/or diabetic individuals, and the presence of T cell infiltrates in the islets of pancreatic specimens from diabetic cadavers.Citation8Citation11 The breakdown of β cell-specific tolerance is complex, involving both genetic and environmental factors, which contribute to dysregulation of mechanisms promoting T cell tolerance.Citation12Citation14 The latter is marked by increased development of type 1 CD4+ and CD8+ effector T cells characterized by the secretion of proinflammatory cytokines such as IFNγ and TNFα.Citation15 The apparent skewed differentiation of naïve β cell-specific T cells towards pathogenic type 1 effectors correlates with reduced numbers and/or function of immunoregulatory T cells (Treg), and/or reduced sensitivity of established type 1 T effectors to Treg-mediated regulation.Citation11,Citation16Citation21 A number of subsets of Treg have been identified which are defined by the: (i) type of cytokine(s) secreted, (ii) effector function(s) employed to regulate an immune response and (iii) overall potency.Citation22

To date most immunotherapies have focused on reestablishing the functional balance between pathogenic type 1 T effectors and Treg to prevent and/or treat T1D. In the clinic the most promising results have been achieved with non-mitogenic (NM) anti-CD3 antibodies administered to recent onset diabetic patients. β cell mass is maintained in these patients; however, protection is relatively short-lived and is associated with transient depletion of T cells which may lead to recurrent viral infections.Citation23Citation25 Other strategies of immunotherapy have been tested in experimental models and the clinic. Antigen-specific immunotherapies have proven to be effective at preventing overt diabetes in NOD mice and transgenic models of T1D, but clinical findings have largely been disappointing with only recent studies providing cause for optimism.Citation26Citation36 This approach is appealing since administration of β cell antigens or peptides under various conditions can be used to selectively manipulate β cell-specific T cell reactivity, with minimal if any effect on the “normal” function of the immune system. Depending on the protocol, administration of β cell antigen may lead to: (1) T cell deletion or induction of a state of unresponsiveness (e.g., anergy), and/or (2) differentiation and expansion of Treg. Clonal anergy or deletion induced by high dose soluble antigen for instance, is exquisitely specific for those T cells recognizing the injected antigen.Citation37 However, at late stages of disease progression when pathogenic CD4+ and CD8+ T cells recognize multiple autoantigens and epitopes, anergy/deletion of a select pool of T cell clones is typically ineffective.Citation37,Citation38 Accordingly, promoting Treg differentiation and/or expansion has generally been the preferred outcome.Citation38Citation41 Once established, Treg can traffick to the islets and draining pancreatic lymph nodes (PLN) and through secretion of cytokines regulate β cell autoimmunity independent of the antigen-specificity of the pathogenic effector T cells.Citation38Citation42 Nevertheless, the efficacy of antigen-based immunotherapy generally wanes at late pre-clinical and clinical stages of intervention partly reflecting the increased numbers of pathogenic type 1 T effectors, and the need for a sufficiently large frequency of Treg.Citation40,Citation42Citation44 Administration of cytokines to promote differentiation and/or expansion of different subsets of Treg has also proven to be effective in preventing overt diabetes in NOD mice. For instance, ongoing β cell autoimmunity is suppressed in NOD mice treated with recombinant IL-4 and IL-10 and the subsequent induction of IL-4 and IL-10 secreting Treg, respectively.Citation45,Citation46 In addition, diabetes is prevented in NOD mice receiving IL-2-antibody complexes which promotes expansion of highly potent Treg expressing the transcription factor FoxP3 (FoxP3+Treg).Citation47,Citation48 However, the pleiotropic effects of cytokines administered systemically are an important concern, especially if the cytokines need to be administered long-term to maintain protection.

Genetic vaccines offer a strategy to enhance the efficacy of antigens, cytokines and other immunomodulatory proteins used to reestablish T cell self-tolerance. Transfer of genes obviates the need to express, purify and store recombinant proteins. Furthermore, genetic vaccination enables greater flexibility in manipulating the nature of a T cell response, in addition to directly modifying in vivo the “tolerogenicity” of the target tissue (e.g., β cells). We will review the most studied approaches of genetic vaccination used to prevent and/or suppress β cell autoimmunity, in addition to highlighting the respective strengths and weaknesses of these strategies.

Clinial Scenarios for Immunotherapy in T1D

There are three general clinical scenarios in which immunotherapy can be applied to suppress β cell autoimmunity and reestablish tolerance within the T cell compartment.Citation41,Citation43,Citation49Citation51 Firstly, immunotherapy can be used to prevent the onset of clinical diabetes in at risk individuals. These individuals are identified by detection of autoantibodies specific for various islet and β cell antigens in serum, in addition to altered insulin responses upon glucose challenge. Secondly, immunotherapy can be applied for the purpose of rescuing residual β cell mass in recent onset and long-term diabetic subjects. At the time of clinical diagnosis a sufficient amount of functional β cell mass persists so that remission of diabetes may be induced if islet inflammation is suppressed.Citation50 Furthermore, indirect evidence suggests that protection of even minimal β cell mass in chronic diabetic patients can result in more efficient glycemic control.Citation52 Finally, immunotherapy can be applied in the context of β cell replacement in chronic diabetic individuals. Recently islet transplantation has proven to be a feasible strategy to provide a “cure” for chronic diabetic patients.Citation51 However, long-term survival of islet grafts depends on persistent tolerance within the pool of β cell-specific T cells. Similarly, efforts to promote β cell regeneration/expansion in vivo are only possible with suppression of the diabetogenic response.

Importantly, the efficacy of a given immunotherapy to suppress β cell autoimmunity is dictated by the number of pathogenic type 1 effector T cells present, and the overall proinflammatory milieu that is established in the islets at the time of intervention. In this regard the most stringent conditions are expected at late preclinical and clinical stages of disease progression.

Genetic Vaccination to Manipulate β cell-specific T Cell Reactivity

To date the use of genetic vaccines to suppress β cell autoimmunity has been studied largely in NOD mice and murine transgenic models of T1D; only recently has this approach been assessed in the clinic. In general, two strategies of genetic vaccination have been studied in depth; namely plasmid DNA (pDNA)- and viral vector-based vaccines ( and ). Recently, a third genetic approach entailing the use of antisense oligonucleotides (AS-ODN) has also proven to be effective for manipulating β cell autoimmunity.

Application of pDNA vaccination to induce β cell-specific T cell tolerance.

pDNA vaccines have been mostly studied for infectious diseases and cancer, with more recent efforts focusing on autoimmunity.Citation53Citation55 Intramuscular (i.m.) injection of soluble or “naked” pDNA results in significant levels of protein expression of the encoded transgene that may persist for 6 weeks or longer.Citation53,Citation56 pDNA vaccines are considered to be safe, in that pDNA fail to integrate into the host genome, exhibit limited immunogenicity, and are well tolerated in the clinic.Citation53,Citation56 From a production standpoint, pDNA are readily manufactured and stored.Citation57 However, the in vivo transfection frequency of pDNA is low, and different cell types are transfected which may lead to varying levels of transgene expression.Citation53Citation56 Different strategies of delivery have been used to increase the efficiency of pDNA transfection. Transfection is markedly enhanced via “gene gun” vaccination for instance, which involves bombardment of the epidermis of the skin with pDNA-coated gold particles.Citation58Citation60 pDNA complexed with cationic polymers or liposomes or the use of electroporation have also been used to increase transfection efficiency.Citation61,Citation62 Nevertheless, pDNA-induced antibody or T cell responses specific for foreign or tumor antigens have generally been weak and/or transient in human subjects.Citation53,Citation54 The latter, however, may in fact be beneficial for preventing and treating autoimmunity, where exacerbating an ongoing pathogenic response must be avoided.

Distinct approaches of pDNA vaccination have been used to immunoregulate β cell autoimmunity (). For instance, i.m. injection of pDNA encoding CCL4 or CXCL10 to young NOD mice results in the induction of neutralizing antibodies specific for the respective chemokines.Citation63,Citation64 Consequently, T cell trafficking to the islets is blocked and the development of diabetes prevented. This approach, however, is limited by the lack of specificity for the autoimmune response.

Induction of β cell-specific Treg differentiation and/or expansion has typically entailed the use of pDNA encoding anti-inflammatory cytokines, β cell autoantigens or the combination of both. Delivery of a short course of pDNA encoding IL-4 or IL-10 to NOD mice early in the diabetogenic response, results in a transient increase in systemic levels of the respective cytokines and prevention of overt diabetes.Citation65,Citation66 These cytokines influence both the differentiation of type 1 and Treg effectors, and block the activation/maturation of antigen presenting cells (APC) such as dendritic cells (DC) and macrophages. However, when administered at later preclinical stages of the diabetogenic response, the efficacy of IL-4 and IL-10 encoding pDNA is reduced.Citation67Citation69 In this instance, islet infiltration is unaffected and diabetes continues to develop in the treated NOD mice. Failure to suppress β cell autoimmunity under increasingly stringent conditions partly reflects inadequate cytokine levels established in the relevant target tissues, namely the islets and draining PLN. The relative immunoregulatory potency of IL-4 and IL-10 may also be a key factor. For example, overt diabetes is prevented in NOD mice at a late preclinical stage of T1D following i.m. injection of pDNA encoding TGFβ1 .Citation70 However, protection is dependent on repeated pDNA injections raising the concern that elevated levels of systemic TGFβ1 long-term may impair normal immune function. An alternative cytokine-based strategy has been to neutralize a given proinflammatory cytokine by administration of pDNA encoding the corresponding soluble receptor. Vaccination with pDNA expressing a soluble fusion molecule consisting of the IFNγ receptor (IFNγR) and IgG-Fc domain prevents islet infiltration and autoimmune diabetes induced by multiple low dose injections of streptozotocin (STZ) in NOD mice.Citation71 However, systemic and persistent expression of the IFNγR-Ig fusion molecule may again impair protective type 1 T cell-mediated immunity specific for pathogens.

Delivery of pDNA encoding β cell autoantigens has proven to be effective at selectively blocking β cell autoimmunity. pDNA encoding insulin B chain, proinsulin, glutamic acid decarboxylase 65 (GAD65) and heat shock protein 60 (HSP60) suppress autoimmunity at various stages of disease progression in NOD mice.Citation67Citation69,Citation72Citation77 A number of factors, however, impact the efficacy of pDNA-mediated β cell specific tolerance. The context of β cell autoantigen expression is a key parameter determining overall efficacy. Induction of Treg by pDNA encoding antigens that are intracellularly expressed is dependent on direct transfection of APC and/or cross-presentation by professional APC of antigen derived from transfected cells, such as myocytes in the case of i.m. injection of pDNA.Citation53 Consequently the number of professional APC such as DC which process and present the corresponding epitopes to T cells may be low, thereby limiting the induction of a sufficient pool of β cell-specific Treg. Increased doses and repeated injections of pDNA may enhance efficacy.Citation75 An alternative approach has been to engineer β cell autoantigens that are secreted.Citation67,Citation78,Citation79 The frequency of GAD65-specific Th2 cells and subsequent diabetes prevention are markedly increased in NOD mice injected i.m. with pDNA encoding a secreted GAD65-IgFc fusion molecule compared to native GAD65 expressed intracellularly.Citation67 Secretion of a β cell autoantigen by pDNA transfected cells ensures widespread distribution in vivo, and in turn an increased frequency of APC that endocytose, process and present the corresponding epitopes.

Administration of pDNA encoding a β cell autoantigen alone has proven to be insufficient at suppressing islet inflammation at later stages of disease progression; in fact β cell autoimmunity may be exacerbated.Citation67,Citation72 Accordingly, one approach has been to co-administer pDNA encoding antigen and anti-inflammatory cytokines to “shape” the nature of the T cell response.Citation67Citation69,Citation80 Intramuscular injection of pDNA encoding GAD65-IgFc and IL-10 suppresses β cell autoimmunity and protects syngeneic islet grafts in diabetic NOD recipients.Citation68,Citation69 In this case co-injection of pDNA encoding IL-10 enhances differentiation of GAD65-specific IL-10-secreting and FoxP3-expressing Treg, and may also potentiate protection by downregulating pathogenic effector T cells and islet resident APC.Citation68,Citation69 Co-injection of pDNA encoding a modified CD80 molecule that binds to CTLA-4-only (mCD80) has also been used to enhance β cell-specific Treg reactivity and prevent diabetes in young NOD mice.Citation81 The site and mode of vaccination also influences the nature of the T cell response elicited by the pDNA encoded β cell autoantigen. For instance, i.m. injection of pDNA results in preferential induction of type 1 effector T cells which can exacerbate β cell autoimmunity in NOD mice.Citation72 Differentiation of type 1 T effectors is partly attributed to CpG motifs found in the vector backbone that bind Toll-like receptor 9 and promote a proinflammatory response.Citation53,Citation56,Citation57 On the other hand, delivery of pDNA to the epidermis via gene gun results in preferential induction of IL-4-secreting Th2 cells independent of CpG motifs.Citation58Citation60 Currently it is unclear why Th2 cell differentiation is selectively promoted but may be due to the tolerogenic properties of epidermal Langerhans cells.Citation82 Similarly, the tolerogenic nature of mucosal tissues can be exploited to manipulate T cell reactivity by delivering pDNA via intranasal or oral routes.Citation76,Citation83 In this way induction/expansion of Treg versus pathogenic type 1 T effectors can be further enhanced.

Promising results have been obtained in a recent phase I/II randomized, dose escalation trial in which diabetic patients receive weekly i.m. injections of pDNA encoding full-length human proinsulin.Citation84 Notably, β cell function as determined by insulin C-peptide levels is maintained over a 12 month period in patients vaccinated with the pDNA encoding proinsulin, leading to improved glycemic control compared to subjects receiving the placebo control. The proinsulin encoding pDNA vaccine is well tolerated and efficacy correlates with reduced anti-insulin antibody titers.Citation84

Viral vector-based vaccination: the use of recombinant adeno-associated virus vectors to induce T cell tolerance.

The majority of studies using viral vector-based vaccines have focused on immunity to infectious pathogens and tumor antigens, although this approach has been employed for prevention and treatment of T1D experimentally ().Citation85 The key advantage of this approach relative to pDNA vaccination is that viral vector-based vaccines typically transduce cells with greater efficiency in vivo. This can result in more robust expression levels of the encoded transgene, and a broader range of tissues (e.g., islets) that can be targeted in vivo. On the other hand, vector toxicity to transduced tissue and vector-specific immunity are key concerns. For instance, the efficacy of replication-defective adenovirus (Ad) vectors is reduced by pre-existing immunity to capsid proteins used to package the recombinants, which in turn affects levels and persistence of transgene expression and limits repeated injection of the recombinant.Citation86 In this regard, recombinant adeno-associated virus (rAAV) vectors have garnered a great deal of interest as an efficient and safe gene transfer platform.

rAAV vectors are highly amenable for gene delivery for a number of reasons. rAAV vectors transduce both dividing and nondividing cells, and exhibit broad tissue tropism with minimal toxicity that leads to long-term transgene expression in vivo without significant immunogenicity.Citation87,Citation88 Furthermore, the risk of genomic insertion and insertional mutagenesis is minimal since rAAV persists as nonintegrating circular monomers or concatemers in the nucleus.Citation89 Clinical studies using rAAV-mediated gene transfer to complement genetic disorders have generated promising results.Citation90 Moreover, improved methods to engineer and produce packaged rAAV coupled with the availability of multiple serotypes to manipulate the immunogenicity of the recombinants enhance clinical application of rAAV-based gene transfer.Citation87,Citation88 The development of double-stranded (ds) rAAV vectors has further improved the approach. Upon delivery, traditional single-stranded (ss) rAAV vectors become transcriptionally active upon conversion to a double stranded DNA template, which results in a slow onset of transgene expression.Citation91 The use of dsAAV vectors eliminates this rate limiting step to accelerate the onset and increase the level of transgene expression.Citation92Citation94 Consequently, lower doses of dsAAV versus ssAAV can be delivered to achieve sufficient levels of transgene expression.

rAAV vectors have been applied in multiple ways to block β cell autoimmunity in NOD mice and other models of T1D (). Intramuscular injection of rAAV1 or rAAV2 vectors and systemic expression of β cell autoantigens (e.g., proinsulin, GAD65),Citation95Citation97 and cytokines (e.g., IL-10),Citation98Citation100 suppresses ongoing β cell autoimmunity at both early and late preclinical stages, and prevents overt diabetes in NOD mice via induction of Treg. Furthermore, i.m. delivery of a rAAV vector expressing IL-10 protects syngeneic islet grafts implanted into diabetic NOD recipients, demonstrating that the approach is robust even at clinical stages of T1D.Citation101 Established β cell autoimmunity in NOD mice is also suppressed by i.m. delivery of rAAV encoding anti-inflammatory molecules such as human α1-antitrypsin (AAT)Citation102 a serine protease inhibitor and heme oxygenase-1 (HO-1)Citation103 a stress-response enzyme that catalyzes the degradation of heme to free iron, carbon monoxide and biliverdin. Here protection is mediated primarily due to the effects of AAT and HO-1 on innate effector cells. Depending on the dose and transgene, the encoded proteins by a given rAAV can be detected several weeks post-injection.Citation95Citation103 Whether sustained expression of high systemic levels of these proteins compromises normal immune function, however, has not been assessed. Inducible promoters to regulate transgene expression can be used to address this potential concern. Intramuscular injection of rAAV encoding an AAT transgene driven by a tetracycline/doxycycline inducible promoter results in increased AAT expression and suppression of collagen-induced arthritis when mice are fed doxycycline containing chow.Citation104 Notably, the level and length of time of gene expression can be effectively manipulated with an inducible promoter so that tolerance can be established and maintained in a safe manner.

A major feature of rAAV-based vaccination is the ability to directly modify the tolerogenicity of β cells in vivo in a cell-specific manner. In this way, possible complications associated with systemic expression of an immunoregulatory molecule are obviated. In addition, direct expression of a given protein in the islets may more readily establish immunotherapeutic levels that otherwise are not attained via a systemic route. The latter may also reduce the required dose of rAAV thereby minimizing the possibility of inducing immunity to the recombinant. Studies have shown employing rAAV encoding green fluorescent protein (GFP) that the efficiency of in vivo transduction of pancreatic tissue is influenced by the serotype of the capsid proteins used for packaging, and the route of rAAV delivery. For instance, rAAV8 vector is highly efficient at transducing murine β cells and acinar cells of the exocrine pancreas when administered via i.v. or intraperitoneal (i.p.) routes, whereas rAAV6 vector is the preferred choice for pancreatic intraductal infusion.Citation105 Importantly, rAAV transduction has no effect on β cell function.Citation105Citation108 It is noteworthy that studies of viral capsid protein structure and the corresponding receptors have led to the engineering of tissue-specific capsids. Random peptide ligand libraries have been used to generate AAV capsid proteins specific for tissues previously resistant to rAAV infection.Citation110Citation112 Furthermore, pseudotyped rAAV have been established in which relevant amino acid sequences from different capsid proteins are swapped to create a tissue-specific chimeric recombinant.Citation113Citation115 These strategies may lead to the future development of capsid proteins that promote efficient, “β cell-only” transduction by rAAV vectors.

Currently since rAAV serotypes that efficiently transduce β cells also transduce other tissues, it is necessary to engineer rAAV vectors with an appropriate promoter to target transgene expression in a tissue-specific manner. Here, the use of an insulin II promoter (IP) has proven to be highly effective for tightly-regulated and stable β cell-specific expression of rAAV encoded transgenes.Citation105,Citation116 Evidence that T1D can be manipulated by targeting β cells in vivo is provided by a study in which dsAAV8 recombinants encoding IL-4 and IL-10 transgenes driven by a mouse IP (mIP) were administered i.p. to young NOD mice.Citation116 Diabetes is prevented in NOD mice receiving dsAAV8-mIP-IL4, which correlates with reduced islet infiltration and an increase in FoxP3+Treg in the periphery. Interestingly, no effect on β cell autoimmunity is detected in dsAAV8-mIP-IL10-treated NOD mice. These observations suggest that local versus systemic expression of a cytokine can have markedly different effects on β cell autoimmunity. For instance, in contrast to β cell-specific expression, rAAV-driven systemic expression of IL-10 but not IL-4 protects NOD mice from diabetes.Citation98 Importantly, the above study provides proof-of-principle that rAAV can be used to modulate the tolerogenicity of β cells in vivo. Whether this strategy is sufficiently robust under more stringent conditions (e.g., late preclinical or clinical stages of T1D) still needs to be determined. In addition, the efficacy of other anti-inflammatory cytokines and/or immunomodulatory molecules need to be tested. It is noteworthy that i.p. injection of dsAAV8 carrying a mIP-driven transgene encoding glucagon-like peptide-1 blocks autoimmune diabetes induced by STZ in BALB/c mice.Citation117

Finally, rAAV vector-based vaccination may also be applied for genetically modifying islet grafts ex vivo for the purpose of inducing transplantation tolerance. Feasibility for this general approach has been provided by numerous studies using Ad vectors (). Genes encoding cytokines, anti-inflammatory and anti-apoptotic proteins and molecules to block T cell co-stimulation (e.g., CTLA-4Ig) have been successfully used to increase islet graft survival.Citation118 Although enhanced, islet graft survival is nevertheless transient due in part to low transduction efficiencies and the immunogenicity of Ad vectors. Accordingly, dsAAV vectors are well suited for modifying the tolerogenicity of islet grafts due to limited immunogenicity and rapid transgene expression. dsAAV packaged in serotypes 2, 6 and 8 efficiently transduce human islets without impairing β cell function.Citation109

Application of antisense therapy to induce T cell tolerance.

Transfer of genes encoding autoantigens and immunomodulatory proteins has been the predominate approach to genetically manipulate autoimmunity in general and T1D specifically. An alternative strategy, however, is to block expression of relevant genes by targeting RNA. A number of different approaches including ribozymes, DNAzymes, aptamers and AS-ODN have been used to mediate “antisense therapy”.Citation119,Citation120 Of these approaches, the use of AS-ODN is arguably the most direct therapeutic strategy and multiple clinical trials testing AS-ODN in for example hematology, oncology and neuromuscular diseases are ongoing.Citation120 AS-ODN are single-stranded DNA molecules designed to specifically hybridize to the complementary RNA. Upon binding, AS-ODN block the function of mRNA by altering splicing events, inhibiting protein translation by influencing ribosome assembly, and/or eliciting endogenous RNase H enzymes.Citation120

AS-ODN have been successfully used to modify the stimulatory capacity of DC either ex vivo or in vivo and in this way suppress β cell autoimmunity in NOD mice. DC uptake of AS-ODN specific for CD40, CD80 and CD86 blocks expression of these co-stimulatory molecules and establishes a robust tolerogenic phenotype.Citation121 Diabetes is prevented in NOD mice following a single injection of bone marrow-derived DC treated with AS-ODN, and protection correlates with an increase in Treg.Citation121 Currently, a phase I clinical trial is underway to test the safety of ex vivo expanded, autologous DC treated with AS-ODN and injected into diabetic patients.Citation122 The potential of antisense therapy to treat T1D has been further demonstrated in a study examining the efficacy of microspheres containing CD40, CD80 and CD86 AS-ODN injected into recent onset diabetic NOD mice.Citation122 Here the microspheres were delivered at a site anatomically proximal to the PLN in an attempt to enhance targeting of the relevant pool of DC. The AS-ODN induced an increased frequency of FoxP3+Treg and diabetes was in fact reversed in some NOD mice.Citation122 Although the efficiency of in vivo uptake of AS-ODN by DC and selective targeting of tissue-specific DC are key issues that still need to be resolved, the above findings provide evidence that this strategy can be effective even under the most stringent of treatment conditions.

Concluding Remarks

Preclinical studies provide evidence indicating that genetic vaccination in general and specifically pDNA- and AAV vector-based vaccines and AS-ODN are effective at reestablishing β cell-specific T cell tolerance. Each of these strategies has key strengths. pDNA vaccination offers a relatively facile and safe strategy to express proteins and modulate β cell autoimmunity systemically. In addition the nature of the T cell response can be readily manipulated by co-delivery of pDNA encoding antigen, cytokines and/or anti-inflammatory modulators. The recent phase I/II trial studying administration of proinsulin encoding pDNA to diabetic patientsCitation84 and encouraging findings from a phase II trial in which multiple sclerosis patients were treated with myelin basic protein expressing pDNA,Citation123 provide evidence suggesting that pDNA vaccination may indeed be effective to manipulate T cell-mediated autoimmunity in the clinic. rAAV vector-based vaccination on the other hand offers an approach to directly modify and enhance the tolerogenicity of β cells in vivo and ex vivo. dsAAV vectors can be used to express immunoregulatory proteins specifically in β cells in vivo by choosing: (i) the appropriate route of delivery, (ii) capsid proteins that preferentially transduce islets and (iii) promoters which selectively drive transgene expression in β cells. Anti-sense therapy via AS-ODN provides a strategy to alter the phenotype and effector function of APC and possibly T cells in the periphery (e.g., PLN). Whether a given strategy of genetic vaccination alone is sufficient to establish long-term protection in patients, especially at late preclinical and clinical stages of T1D is a question that still needs to be addressed. However, genetic vaccination may also prove to be effective in the context of a combinatorial immunotherapy. In this regard, two potential scenarios can be envisioned.

In the first scenario, different genetic vaccine strategies are combined, similar to heterologous prime-boost vaccination protocols that exploit the properties of distinct vaccines to induce immunity to pathogens.Citation124 One possible approach for example is to induce β cell-specific Treg via autoantigen-encoding pDNA and quench the inflammatory milieu of the islets with dsAAV encoding an anti-inflammatory molecule(s) (e.g., IL-4, AAT, HO-1). This combination may reduce the stringency needed to suppress β cell autoimmunity at later preclinical or clinical stages of T1D in terms of the number and/or type of pDNA-induced β cell-specific Treg, and/or the efficiency of islet transduction and the level of transgene expression by the rAAV vector. Similarly, a more robust β cell-specific Treg response may be elicited by inducing tolerogenic DC in vivo via encapsulated AS-ODN targeting co-stimulatory molecule expression coupled with pDNA- or rAAV vector-encoded β cell autoantigen.

In the second scenario, genetic vaccination can be paired with other “nongenetic-based” strategies of immunotherapy. A recent study assessed the efficacy of i.m. injected pDNA encoding GAD65 combined with i.v. injected NM anti-CD3 antibody in a transgenic model of T1D in which the lymphocytic choriomeningitis virus (LCMV) glycoprotein is a neo-autoantigen expressed by β cells.Citation125 Diabetes is induced in this model by LCMV infection. Notably, the combination immunotherapy reverses diabetes in a greater frequency of recent onset mice relative to either approach alone. This synergy correlates with an increased GAD65-specific Treg response induced by the pDNA-GAD65 and NM anti-CD3 antibody combined treatment. The NM anti-CD3 antibody establishes conditions permissive for GAD65-specific Treg differentiation/expansion by in part depleting pathogenic type 1 T effectors and reducing the overall proinflammatory milieu in the islets and PLN.Citation126,Citation127 These findings demonstrate the potential potency of a combinatorial immunotherapy, and establish rationale for combining genetic vaccines with approaches based on administration of other antibodies (e.g., anti-CD20) and/or immunomodulatory proteins (e.g., vitamin D).

In conclusion, genetic vaccination can be used to manipulate the diabetogenic response either systemically, and/or by directly modifying the tolerogenicity of β cells. The inherent flexibility of the approach provides immense potential for clinical application either as a stand alone or combinatorial immunotherapy. Continued preclinical and clinical studies, however, are needed to meet this potential. How treatment parameters (e.g., dose, route of administration) and antigen-specificity of pDNA vaccination influence β cell-specific T cell reactivity at various stages of disease progression for instance, need to be assessed in patients. In addition further preclinical development of dsAAV vectors to improve selective targeting of β cells in vivo, and identifying the most effective immunoregulatory proteins that suppress inflammation in the islets is required. Finally, improved in vivo targeting of and uptake by specific cell types (e.g., DC) is needed to enhance the clinical application of AS-ODN microspheres.

Abbreviations

AAV=

adeno-associated virus

Ad=

adenovirus

APC=

antigen presenting cells

AAT=

α1-antitrypsin

AS-ODN=

antisense oligonucleotides

DC=

dendritic cell

ds=

double stranded

GAD65=

glutamic acid decarboxylase 65

GFP=

green fluorescent protein

HO-1=

heme oxygenase-1

Treg=

immunoregulatory T cells

i.m.=

intramuscular

i.p.=

intraperitoneal

i.v.=

intravenous

LCMV=

lymphocytic choriomeningitis virus

mIP=

mouse insulin promoter

NM=

nonmitogenic

NOD=

nonobese diabetic

PLN=

pancreatic lymph nodes

pDNA=

plasmid DNA

ss=

single stranded

STZ=

streptozotocin

T1D=

type 1 diabetes

Figures and Tables

Table 1 Approaches of pDNA vaccination for reestablishing β cell tolerance

Table 2 Viral-based vectors primarily used in the treatment of Type 1 diabetes

Acknowledgements

This work was supported by funding from the National Institutes of Health (NIH) (R01AI058014 and R01DK081585 (R.T.)). M.C.J. was supported by a NIH training grant (5T32 AI07273).

References

  • Anderson MS, Bluestone JA. The NOD mouse: a model of immune dysregulation. Annu Rev Immunol 2005; 23:447 - 485
  • Bach JF. Insulin-dependent diabetes mellitus as an autoimmune disease. Endocr Rev 1994; 15:516 - 542
  • Eisenbarth GS. Prediction of type 1 diabetes: the natural history of the prediabetic period. Adv Exp Med Biol 2004; 552:268 - 290
  • Tisch R, McDevitt HO. Insulin-dependent diabetes mellitus. Cell 1996; 85:291 - 297
  • Bendelac A, Carnaud C, Boitard C, Bach JF. Syngeneic transfer of autoimmune diabetes from diabetic NOD mice to healthy neonates. Requirement for both L3T4+ and Lyt-2+ T cells. J Exp Med 1987; 166:823 - 832
  • Christianson SW, Shultz LD, Leiter EH. Adoptive transfer of diabetes into immunodeficient NOD-scid/scid mice. Relative contributions of CD4+ and CD8+ T-cells from diabetic versus prediabetic NOD.NON-Thy-1a donors. Diabetes 1993; 42:44 - 55
  • Miller BJ, Appel MC, O'Neil JJ, Wicker LS. Both the Lyt-2+ and L3T4+ T cell subsets are required for the transfer of diabetes in nonobese diabetic mice. J Immunol 1988; 140:52 - 58
  • Standifer NE, Burwell EA, Gersuk VH, Greenbaum CJ, Nepom GT. Changes in autoreactive T cell avidity during type 1 diabetes development. Clin Immunol 2009; 132:312 - 320
  • Velthuis JH, Unger WW, Abreu JR, Duinkerken G, Franken K, Peakman M, et al. Simultaneous detection of circulating autoreactive CD8+ T cells specific for different islet cell-associated epitopes using combinatorial MHC-multimers. Diabetes 2010; [Epub ahead of print]
  • Coppieters KT, von Herrath MG. Histopathology of type 1 diabetes: old paradigms and new insights. Rev Diabet Stud 2009; 6:85 - 96
  • Arif S, Tree TI, Astill TP, Tremble JM, Bishop AJ, Dayan CM, et al. Autoreactive T cell responses show proinflammatory polarization in diabetes but a regulatory phenotype in health. J Clin Invest 2004; 113:451 - 463
  • Todd JA, Wicker LS. Genetic protection from the inflammatory disease type 1 diabetes in humans and animal models. Immunity 2001; 15:387 - 395
  • Sarvetnick N. Etiology of autoimmunity. Immunol Res 2000; 21:357 - 362
  • Benoist C, Mathis D. Autoimmunity provoked by infection: how good is the case for T cell epitope mimicry?. Nat Immunol 2001; 2:797 - 801
  • Tisch R, Wang B. Dysregulation of T peripheral tolerance in type 1 diabetes. Adv Immunol 2008; 100:125 - 149
  • Brusko TM, Wasserfall CH, Clare-Salzler MJ, Schatz DA, Atkinson MA. Functional defects and the influence of age on the frequency of CD4+ CD25+ T-cells in type 1 diabetes. Diabetes 2005; 54:1407 - 1414
  • Fox CJ, Danska JS. IL-4 expression at the onset of islet inflammation predicts nondestructive insulitis in nonobese diabetic mice. J Immunol 1997; 158:2414 - 2424
  • Gregori S, Giarratana N, Smiroldo S, Adorini L. Dynamics of pathogenic and suppressor T cells in autoimmune diabetes development. J Immunol 2003; 171:4040 - 4047
  • Herman AE, Freeman GJ, Mathis D, Benoist C. CD4+CD25+ T regulatory cells dependent on ICOS promote regulation of effector cells in the prediabetic lesion. J Exp Med 2004; 199:1479 - 1489
  • Lindley S, Dayan CM, Bishop A, Roep BO, Peakman M, Tree TI. Defective suppressor function in CD4+CD25+ T-cells from patients with type 1 diabetes. Diabetes 2005; 54:92 - 99
  • Pop SM, Wong CP, Culton DA, Clarke SH, Tisch R. Single cell analysis shows decreasing FoxP3 and TGFbeta1 coexpressing CD4+CD25+ regulatory T cells during autoimmune diabetes. J Exp Med 2005; 201:1333 - 1346
  • Shevach EM. From vanilla to 28 flavors: multiple varieties of T regulatory cells. Immunity 2006; 25:195 - 201
  • Herold KC, Hagopian W, Auger JA, Poumian-Ruiz E, Taylor L, Donaldson D, et al. Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N Engl J Med 2002; 346:1692 - 1698
  • Keymeulen B, Vandemeulebroucke E, Ziegler AG, Mathieu C, Kaufman L, Hale G, et al. Insulin needs after CD3-antibody therapy in new-onset type 1 diabetes. N Engl J Med 2005; 352:2598 - 2608
  • Herold KC, Gitelman SE, Masharani U, Hagopian W, Bisikirska B, Donaldson D, et al. A single course of anti-CD3 monoclonal antibody hOKT3gamma1(Ala-Ala) results in improvement in C-peptide responses and clinical parameters for at least 2 years after onset of type 1 diabetes. Diabetes 2005; 54:1763 - 1769
  • Kaufman DL, Clare-Salzler M, Tian J, Forsthuber T, Ting GS, Robinson P, et al. Spontaneous loss of T-cell tolerance to glutamic acid decarboxylase in murine insulin-dependent diabetes. Nature 1993; 366:69 - 72
  • Tisch R, Yang XD, Singer SM, Liblau RS, Fugger L, McDevitt HO. Immune response to glutamic acid decarboxylase correlates with insulitis in non-obese diabetic mice. Nature 1993; 366:72 - 75
  • Tisch R, Liblau RS, Yang XD, Liblau P, McDevitt HO. Induction of GAD65-specific regulatory T-cells inhibits ongoing autoimmune diabetes in nonobese diabetic mice. Diabetes 1998; 47:894 - 899
  • Fife BT, Guleria I, Gubbels-Bupp M, Eagar TN, Tang Q, Bour-Jordan H, et al. Insulin-induced remission in new onset NOD mice is maintained by the PD-1-PD-L1 pathway. J Exp Med 2006; 203:2737 - 2747
  • Muir A, Peck A, Clare-Salzler M, Song YH, Cornelius J, Luchetta R, et al. Insulin immunization of nonobese diabetic mice induces a protective insulitis characterized by diminished intraislet interferon-gamma transcription. J Clin Invest 1995; 95:628 - 634
  • Coon B, An LL, Whitton JL, von Herrath MG. DNA immunization to prevent autoimmune diabetes. J Clin Invest 1999; 104:189 - 194
  • Harrison LC, Honeyman MC, Steele CE, Stone NL, Sarugeri E, Bonifacio E, et al. Pancreatic beta cell function and immune response to insulin after administration of intranasal insulin to humans at risk for type 1 diabetes. Diabetes Care 2004; 27:2348 - 2355
  • Skyler JS, Krischer JP, Wolfsdorf J, Cowie C, Palmer JP, Greenbaum C, et al. Effects of oral insulin in relatives of patients with type 1 diabetes: The Diabetes Prevention Trial-Type 1. Diabetes Care 2005; 28:1068 - 1076
  • Orban T, Farkas K, Jalahej H, Kis J, Treszl A, Falk B, et al. Autoantigen-specific regulatory T cells induced in patients with type 1 diabetes mellitus by insulin B-chain immunotherapy. J Autoimmun 2010; 34:408 - 415
  • Ludvigsson J, Faresjo M, Hjorth M, Axelsson S, Cheramy M, Pihl M, et al. GAD treatment and insulin secretion in recent-onset type 1 diabetes. N Engl J Med 2008; 359:1909 - 1920
  • Diabetes Prevention Trial-Type 1 Diabetes Study Group. Effects of insulin in relatives of patients with type 1 diabetes mellitus. N Eng J Med 2002; 346:1685 - 1691
  • Liblau RS, Pearson CI, Shokat K, Tisch R, Yang XD, McDevitt HO. High-dose soluble antigen: peripheral T-cell proliferation or apoptosis. Immunol Rev 1994; 142:193 - 208
  • Tisch R, McDevitt HO. Antigen-specific immunotherapy: is it a real possibility to combat T-cell-mediated autoimmunity?. Proc Natl Acad Sci USA 1994; 91:437 - 438
  • Harrison LC, Hafler DA. Antigen-specific therapy for autoimmune disease. Curr Opin Immunol 2000; 12:704 - 711
  • Fousteri G, Bresson D, von Herrath M. Rational development of antigen-specific therapies for type 1 diabetes. Adv Exp Med Biol 2007; 601:313 - 319
  • Luo X, Herold KC, Miller SD. Immunotherapy of Type 1 diabetes: Where are we and where should we be going?. Immunity 2010; 32:488 - 499
  • Wang B, Tisch R. Parameters influencing antigen-specific immunotherapy for Type 1 diabetes. Immunol Res 2008; 42:246 - 258
  • Staeva-Vieira T, Peakman M, von Herrath M. Translational mini-review series on type 1 diabetes: Immune-based therapeutic approaches for type 1 diabetes. Clin Exp Immunol 2007; 148:17 - 31
  • Tian J, Kaufman DL. Antigen-based therapy for the treatment of type 1 diabetes. Diabetes 2009; 58:1939 - 1946
  • Rapoport MJ, Jaramillo A, Zipris D, Lazarus AH, Serreze DV, Leiter EH, et al. Interleukin 4 reverses T cell proliferative unresponsiveness and prevents the onset of diabetes in nonobese diabetic mice. J Exp Med 1993; 178:87 - 99
  • Pennline KJ, Roque-Gaffney E, Monahan M. Recombinant human IL-10 prevents the onset of diabetes in the nonobese diabetic mouse. Clin Immunol Immunopathol 1994; 71:169 - 175
  • Tang Q, Adams JY, Penaranda C, Melli K, Piaggio E, Sgouroudis E, et al. Central role of defective interleukin-2 production in triggering islet autoimmune destruction. Immunity 2008; 28:687 - 697
  • Sakaguchi S. Naturally arising CD4+ regulatory T cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol 2004; 22:531 - 562
  • Haller MJ, Gottlieb PA, Schatz DA. Type 1 diabetes intervention trials 2007: where are we and where are we going?. Curr Opin Endocrinol Diabetes Obes 2007; 14:283 - 287
  • Bougneres PF, Landais P, Boisson C, Carel JC, Frament N, Boitard C, et al. Limited duration of remission of insulin dependency in children with recent overt type I diabetes treated with low-dose cyclosporin. Diabetes 1990; 39:1264 - 1272
  • Ryan EA, Paty BW, Senior PA, Bigam D, Alfadhi E, Kneteman NM, et al. Five-year follow-up after clinical islet transplantation. Diabetes 2005; 54:2060 - 2069
  • Shah SC, Malone JI, Simpson NE. A randomized trial of intensive insulin therapy in newly diagnosed insulin-dependent diabetes mellitus. N Engl J Med 1989; 320:550 - 554
  • Donnelly JJ, Wahren B, Liu MA. DNA vaccines: progress and challenges. J Immunol 2005; 175:633 - 639
  • Lu S, Wang S, Grimes-Serrano JM. Current Progress of DNA vaccine studies in humans. Expert Rev Vaccines 2008; 7:175 - 191
  • Garren H, Steinman L. Fathman CG. DNA vaccination in the treatment of autoimmune disease. Biologic and Gene Therapy of Autoimmune Disease 2000; Basel Karger 203 - 216
  • Abdulhaqq SA, Weiner DB. DNA vaccines: developing new strategies to enhance immune responses. Immunol Res 2008; 42:219 - 232
  • Williams JA, Carnes AE, Hodgson CP. Plasmid DNA vaccine vector design: Impact on efficacy, safety and upstream production. Biotech Adv 2009; 27:353 - 370
  • Feltquate DM, Heaney S, Webster RG, Robinson HL. Different T helper cell types and antibody isotypes generated by saline and gene gun DNA immunization. J Immunol 1997; 158:2278 - 2284
  • Weiss R, Scheiblhofer S, Freund J, Ferreira F, Livey I, Thalhamer J. Gene gun bombardment with gold particles displays a particular Th2-promoting signal that over-rules the Th1-inducing effect of immunostimulatory CpG motifs in DNA vaccines. Vaccine 2002; 20:3148 - 3154
  • Goudy KS, Wang B, Tisch R. Gene gun-mediated DNA vaccination enhances antigen-specific immunotherapy at a late preclinical stage of type 1 diabetes on nonobese diabetic mice. Clin Immunol 2008; 129:49 - 57
  • Ewert KK, Ahmad A, Bouxsein NF, Evans HM, Safinya CR. Non-viral gene delivery with cationic liposome-DNA complexes. Methods Mol Biol 2008; 433:159 - 175
  • Bodles-Brakhop AM, Heller R, Draghia-Akli R. Electroporation for the delivery of DNA-based vaccines and immunotherapeutics: current clinical developments. Mol Ther 2009; 17:585 - 592
  • Shigihara T, Shimada A, Oikawa Y, Yoneyama H, Kanazawa Y, Okubo Y, et al. CXCL10 DNA vaccination prevents spontaneous diabetes through enhanced β cell proliferation in NOD mice. J Immunol 2005; 175:8401 - 8408
  • Meagher C, Arreaza G, Peters A, Strathdee CA, Gilbert PA, Mi QS, et al. CCL4 protects from Type 1 diabetes by altering islet β cell-targeted inflammatory responses. Diabetes 2007; 56:809 - 817
  • Cameron MJ, Strathdee CA, Holmes KD, Arreaza GA, Dekaban GA, Delovitch TL. Biolistic-mediated interleukin 4 gene transfer prevents onset of Type 1 diabetes. Human Gene Ther 2000; 11:1647 - 1656
  • Nitta Y, Tashiro F, Tokui M, Shimada A, Takei I, Tabayashi K, et al. Systemic delivery of interleukin 10 by intramuscular injection of expression plasmid DNA prevents autoimmune diabetes in nonobese diabetic mice. Human Gene Ther 1998; 9:1701 - 1707
  • Tisch R, Wang B, Weaver DJ, Liu B, Bui T, Arthos J, Serreze DV. Antigen-specific mediated suppression of beta cell autoimmunity by plasmid DNA vaccination. J Immunol 2001; 166:2122 - 2132
  • Seifarth C, Pop S, Liu B, Wong CP, Tisch R. More stringent conditions of plasmid DNA vaccination are required to protect grafted versus endogenous islets in nonobese diabetic mice. J Immunol 2003; 171:469 - 476
  • Pop SM, Wong CP, He Q, Wang Y, Wallet MA, Goudy KS, et al. The type and frequency of immunoregulatory CD4+ T-cells govern the efficacy of antigen-specific immunotherapy in nonobese diabetic mice. Diabetes 2007; 56:1395 - 1402
  • Piccirillo CA, Chang Y, Prud'homme GJ. TGFβ1 somatic gene therapy prevents autoimmune disease in nonobese diabetic mice. J Immunol 1998; 161:3950 - 3956
  • Prud'homme GJ, Chang Y. Prevention of autoimmune diabetes by intramuscular gene therapy with a nonviral vector encoding an interferon-gamma receptor/IgG1 fusion protein. Gene Ther 1999; 6:771 - 777
  • Weaver DJ, Liu B, Tisch R. Plasmid DNAs encoding insulin and glutamic acid decarboxylase 65 have distinct effects on the progression of autoimmune diabetes in nonobese diabetic mice. J Immunol 2001; 167:586 - 592
  • Bot A, Smith D, Bot S, Hughes A, Wolfe T, Wang L, et al. Plasmid vaccination with insulin B chain prevents autoimmune diabetes in nonobese diabetic mice. J Immunol 2001; 167:2950 - 2955
  • Urbanek-Ruiz I, Ruiz PJ, Paragas V, Garren H, Steinman L, Fathman CG. Immunization with DNA encoding an immunodominant peptide of insulin prevents diabetes in NOD mice. Clin Immunol 2001; 100:164 - 171
  • Solvason N, Lou YP, Peters W, Evans E, Martinez J, Ramirez U, et al. Improved efficacy of a tolerizing DNA vaccine for reversal of hyperglycemia through enhancement of gene expression and localization to intracellular sites. J Immunol 2008; 181:8298 - 8307
  • Every AL, Kramer DR, Mannering SI, Lew AM, Harrison LC. Intranasal vaccination with proinsulin DNA induces regulatory CD4+ T cells that prevent experimental autoimmune diabetes. J Immunol 2006; 176:4608 - 4615
  • 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
  • Glinka Y, De PR, Croze F, Prud'homme GJ. Regulatory cytokine production stimulated by DNA vaccination against an altered form of glutamic acid decarboxylase 65 in nonobese diabetic mice. J Mol Med 2003; 81:175 - 184
  • Filippova M, Liu J, Escher A. Effects of plasmid DNA injection on cyclophosphamide-accelerated diabetes in NOD mice. DNA Cell Biol 2001; 20:175 - 181
  • Wolfe T, Bot A, Hughes A, Mohrle U, Rodrigo E, Jaume JC, et al. Endogenous expression levels of autoantigens influence success or failure of DNA immunizations to prevent type 1 diabetes: addition of IL-4 increases safety. Eur J Immunol 2002; 32:113 - 121
  • Glinka Y, Chang Y, Prud'homme GJ. Protective regulatory T cell generation in autoimmune diabetes by DNA covaccination with islet antigens and selective CTLA-4 ligand. Mol Ther 2006; 578 - 587
  • Kaplan DH, Jenison MC, Saeland S, Shlomchik WD, Shlomchik MJ. Epidermal langerhans cell-deficient mice develop enhanced contact hypersensitivity. Immunity 2005; 23:611 - 620
  • Li AF, Escher A. Intradermal or oral delivery of GADencoding genetic vaccines suppress type 1 diabetes. DNA Cell Biol 2003; 22:227 - 232
  • Gottlieb P, Colman PG, Kipnes M, Ratner R, Aroda V, Rendell M, et al. Interim results of a phase I/II clinical trial of a DNA plasmid vaccine (BHT-3021) for type 1 diabetes 69th Annual Meeting of the American Diabetes Association June 5–9; 2009 New Orleans LA
  • Draper SJ, Heeney JL. Viruses as vaccine vectors for infectious diseases and cancer. Nature Rev 2010; 8:62 - 73
  • Lasaro MO, Ertl HCJ. New insights on adenovirus as vaccine vectors. Mol Ther 2009; 17:1333 - 1339
  • Daya S, Berns KI. Gene therapy using adeno-associated virus vectors. Clin Micro Rev 2008; 21:583 - 593
  • Gray SJ, Samulski RJ. Optimizing gene delivery vectors for the treatment of heart disease. Expert Opin Biol Ther 2008; 8:911 - 922
  • Smith RH. Adeno-associated virus integration: virus versus vector. Gene Ther 2008; 15:817 - 822
  • Mueller C, Flotte TR. Clinical therapy using recombinant adeno-associated virus vectors. Gene Ther 2008; 15:858 - 863
  • Ferrari FK, Samulski T, Shenk T, Samulski RJ. Secondstrand synthesis is a rate-limiting step for efficient transduction by recombinant adeno-associated virus vectors. J Virol 1996; 70:3227 - 3234
  • Wang Z, Ma HI, Li J, Sun L, Zhang J, Xiao X. Rapid and highly efficient transduction by double-stranded adeno-associated virus vectors in vitro and in vivo. Gene Ther 2003; 10:2105 - 2111
  • McCarty DM, Fu H, Monahan PE, Toulson CE, Naik P, Samulski RJ. Adeno-associated virus terminal repeat (TR) mutant generates self-complementary vectors to overcome the rate-limiting step to transduction in vivo. Gene Ther 2003; 10:2112 - 2118
  • McCarty DM. Self-complementary AAV vectors; advances and applications. Mol Ther 2008; 16:1648 - 1656
  • Han G, Li Y, Wang J, Wang R, Chen G, Song L, et al. Active tolerance induction and prevention of autoimmune diabetes by immunogene therapy using recombinant adenoassociated virus expressing glutamic acid decarboxylase 65 peptide GAD(500-585). J Immunol 2005; 174:4516 - 4524
  • Han G, Wang R, Chen G, Wang J, Xu R, Feng J, et al. Gene delivery GAD 500 autoantigen by AAV serotype 1 prevented diabetes in NOD mice: transduction efficiency do not play important roles. Immunol Lett 2008; 115:110 - 116
  • Jindal RM, Karanam M, Shah R. Prevention of diabetes in the NOD mouse by intra-muscular injection of recombinant adeno-associated virus containing the preproinsulin II gene. Int J Exp Diabetes Res 2001; 2:129 - 138
  • Goudy K, Song S, Wasserfall C, Zhang YC, Kapturczak M, Muir A, et al. Adeno-associated virus vector-mediated IL-10 gene delivery prevents type 1 diabetes in NOD mice. Proc Natl Acad Sci USA 2001; 98:13913 - 13918
  • Goudy KS, Burkhardt BR, Wasserfall C, Song S, Campbell-Thompson ML, Brusko T, et al. Systemic overexpression of IL-10 induces CD4+CD25+ cell populations in vivo and ameliorates type 1 diabetes in nonobese diabetic mice in a dose-dependent fashion. J Immunol 2003; 171:2270 - 2278
  • Yang Z, Chen M, Wu R, Fialkow LB, Bromberg JS, McDuffie M, et al. Suppression of autoimmune diabetes by viral IL-10 gene transfer. J Immunol 2002; 168:6479 - 6485
  • Zhang YC, Pileggi A, Agarwal A, Molano RD, Powers M, Brusko T, et al. Adeno-associated virus-mediated IL-10 gene therapy inhibits diabetes recurrence in syngeneic islet cell transplantation of NOD mice. Diabetes 2003; 52:708 - 716
  • Song S, Goudy K, Campbell-Thompson M, Wasserfall C, Scott-Jorgensen M, Wang J, et al. Recombinant adeno-associated virus-mediated alpha-1 antitrypsin gene therapy prevents type I diabetes in NOD mice. Gene Ther 2004; 11:181 - 186
  • Hu CM, Lin HH, Chiang MT, Chang PF, Chau LY. Systemic expression of heme oxygenase-1 ameliorates type 1 diabetes in NOD mice. Diabetes 2007; 56:1240 - 1247
  • Grimstein C, Choi YK, Satoh M, Lu Y, Wang X, Campbell-Thompson M, et al. Combination of alpha-1 antitrypsin and doxycycline suppresses collagen-induced arthritis. J Gene Med 2010; 12:35 - 44
  • Wang Z, Zhu T, Rehman KK, Bertera S, Zhang J, Chen C, et al. Widespread and stable pancreatic gene transfer by adeno-associated virus vectors via different routes. Diabetes 2006; 55:875 - 884
  • Flotte T, Agarwal A, Wang J, Song S, Fenjves ES, Inverardi L, et al. Efficient ex vivo transduction of pancreatic islet cells with recombinant adeno-associated virus vectors. Diabetes 2001; 50:515 - 520
  • Kapturczak M, Zolotukhin S, Cross J, Pileggi A, Molano RD, Jorgensen M, et al. Transduction of human and mouse pancreatic islet cells using a bicistronic recombinant adeno-associated viral vector. Mol Ther 2002; 5:154 - 160
  • Prasad KM, Yang Z, Bleich D, Nadler JL. Adeno-associated virus vector mediated gene transfer to pancreatic beta cells. Gene Ther 2000; 7:1553 - 1561
  • Rehman KK, Wang Z, Bottino R, Balamurugan AN, Trucco M, Li J, et al. Efficient gene delivery to human and rodent islets with double-stranded (ds) AAV-based vectors. Gene Ther 2005; 12:1313 - 1323
  • Michelfelder S, Kohlschutter J, Skorupa A, Pfennings S, Muller O, Kleinschmidt JA, et al. Successful expansion but not complete restriction of tropism of adeno-associated virus by in vivo biopanning of random virus display peptide libraries. PLoS One 2009; 4:5122
  • Muller OJ, Kaul F, Weitzman MD, Pasqualini R, Arap W, Kleinschmidt JA, et al. Random peptide libraries displayed on adeno-associated virus to select for targeted gene therapy vectors. Nat Biotechnol 2003; 21:1040 - 1046
  • Work LM, Buning H, Hunt E, Nicklin SA, Denby L, Britton N, et al. Vascular bed-targeted in vivo gene delivery using tropism-modified adeno-associated viruses. Mol Ther 2006; 13:683 - 693
  • Dodiya HB, Bjorklund T, Stansell J 3rd, Mandel RJ, Kirik D, Kordower JH. Differential transduction following basal ganglia administration of distinct pseudotyped AAV capsid serotypes in nonhuman primates. Mol Ther 2010; 18:579 - 587
  • Grimm D, Zhou S, Nakai H, Thomas CE, Storm TA, Fuess S, et al. Preclinical in vivo evaluation of pseudotyped adeno-associated virus vectors for liver gene therapy. Blood 2003; 102:2412 - 2419
  • Rebuffat A, Harding CO, Ding Z, Thony B. Comparison of adeno-associated virus pseudotype 1, 2 and 8 vectors administered by intramuscular injection in the treatment of murine phenylketonuria. Hum Gene Ther 2010; 21:463 - 477
  • Rehman KK, Trucco M, Wang Z, Xiao X, Robbins PD. AAV8-mediated gene transfer of interleukin-4 to endogenous beta-cells prevents the onset of diabetes in NOD mice. Mol Ther 2008; 16:1409 - 1416
  • Riedel MJ, Gaddy DF, Asadi A, Robbins PD, Kieffer TJ. DsAAV8-mediated expression of glucagon-like peptide-1 in pancreatic beta-cells ameliorates streptozotocin-induced diabetes. Gene Ther 2010; 17:171 - 180
  • Giannoukakis N, Trucco M. Gene therapy for type 1 diabetes. Am J Ther 2005; 12:512 - 528
  • Crooke ST. Progress in antisense technology. Annu Rev Med 2004; 55:61 - 95
  • Bennett CF, Swayze EE. RNA targeting therapeutics: molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annu Rev Pharmacol 2010; 50:259 - 293
  • Machen J, Harnaha J, Lakomy R, Styche A, Trucco M, Giannoukakis N. Antisense oligonucleotides downregulating costimulation confer diabetes-preventive properties to nonobese diabetic mouse dendritic cells. J Immunol 2004; 173:4331 - 4341
  • Phillips B, Nylander K, Harnaha J, Machen J, Lakomy R, Styche A, et al. A microsphere-based vaccine prevents and reverses new-onset autoimmune diabetes. Diabetes 2008; 57:1544 - 1555
  • Garren H. A DNA vaccine for multiple sclerosis. Expert Opin Biol Ther 2008; 8:1539 - 1549
  • Ranasinghe C, Ramshaw IA. Genetic heterologous prime-boost vaccination strategies for improved systemic and mucosal immunity. Expert Rev Vaccines 2009; 8:1171 - 1181
  • Bresson D, Fradkin M, Manenkova Y, Rottembourg D, von Herrath M. Genetic-induced variations in the GAD65 T cell repertoire governs efficacy of anti-CD3/GAD65 combination therapy in new-onset type 1 diabetes. Mol Ther 2010; 18:307 - 316
  • Belghith M, Bluestone JA, Barriot S, Megret J, Bach JF, Chatenoud L. TGF-beta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes. Nat Med 2003; 9:1202 - 1208
  • Chatenoud L, Primo J, Bach JF. CD3 antibody-induced dominant self tolerance in overtly diabetic NOD mice. J Immunol 1997; 158:2947 - 2954
  • Ko KS, Lee M, Koh JJ, Kim SW. Combined administration of plasmids encoding IL-4 and IL-10 prevents the development of autoimmune diabetes in nonobese diabetic miice. Mol Ther 2001; 4:313 - 316
  • Balasa B, Boehm BO, Fortnagel A, Karges W, Van Gunst K, Jung N, et al. Vaccination with glutamic acid decarboxylase plasmid DNA protects mice from spontaneous autoimmune diabetes and B7/CD28 costimulation circumvents that protection. Clin Immunol 2001; 99:241 - 252
  • Glinka Y, Pooter R, Croze F, Prud'homme GJ. Regulatory cytokine production stimulated by DNA vaccination against an altered form of glutamic acid decarboxylase 65 in nonobese diabetic mice. J Mol Med 2003; 81:175 - 184
  • Prud'homme GJ, Chang Y, Li X. Immunoinhibitory DNA vaccine protects against autoimmune diabetes through cDNA encoding a selective CTLA-4 (CD152) ligand. Human Gene Ther 2002; 13:395 - 406
  • Li A, Ojogho O, Franco E, Baron P, Iwaki Y, Escher A. Pro-apoptotic DNA vaccination ameliorates new onset of autoimmune diabetes in NOD mice and induces foxp3+ regulatory T cells in vitro. Vaccine 2006; 24:5036 - 5046
  • Yamada K, Moriyama H, Okumachi Y, Arai T, Kameno M, Kishi M, et al. Intravenous administration of proinsulin 1 or 2-expressing fiber-mutant recombinant adenovirus vector protects against the development of diabetes in NOD mice. Ann NY Acad Sci 2008; 1150:183 - 186
  • Fernandes JR, Duvivier-Kali VF, Keegan M, Hollister-Lock J, Omer A, Su S, et al. Transplantation of islets transduced with CTLA4-Ig and TGFbeta using adenovirus and lentivirus vectors. Transpl Immunol 2004; 13:191 - 200
  • Park L, Lee E, Lee S, Lim M, Hong H, Shin G, et al. TGFbeta plasmid construction and delivery for the prevention of type 1 diabetes. Ann N Y Acad Sci 2008; 1150:177 - 182
  • Luo X, Yang H, Kim IS, Saint-Hillaire F, Thomas DA, De BP, et al. Systemic transforming growth factor-beta1 gene therapy induced FoxP3+ regulatory cells, restores self-tolerance, and facilitates regeneration of beta cell function in overtly diabetic nonobese diabetic mice. Transplantation 2005; 79:1091 - 1096
  • Sakata M, Yasuda H, Moriyama H, Yamada K, Kotani R, Kurohara M, et al. Prevention of recurrent but not spontaneous autoimmune diabetes by transplanted NOD islets adenovirally transduced with immunomodulating molecules. Diabetes Res Clin Pract 2008; 80:352 - 359
  • Yasuda H, Nagata M, Arisawa K, Yoshida R, Fujihira K, Okamoto N, et al. Local expression of immunoregulatory IL-12p40 gene prolonged syngeneic islet graft survival in diabetic NOD mice. J Clin Invest 1998; 102:1807 - 1814
  • Smith DK, Korbutt GS, Suarez-Pinzon WL, Kao D, Rajotte RV, Elliott JF. Interleukin-4 or interleukin-10 expressed from adenovirus-transduced syngeneic islet grafts fails to prevent beta cell destruction in diabetic NOD mice. Transplantation 1997; 64:1040 - 1049
  • Cameron MJ, Arreaza GA, Waldhauser L, Gauldie J, Delovitch TL. Immunotherapy of spontaneous type 1 diabetes in nonobese diabetic mice by systemic interleukin-4 treatment employing adenovirus vector-mediated gene transfer. Gene Ther 2000; 7:1840 - 1846
  • Alexander AM, Crawford M, Bertera S, Rudert WA, Takikawa O, Robbins PD, et al. Indoleamine 2,3-dioxygenase expression in transplanted NOD Islets prolongs graft survival after adoptive transfer of diabetogenic splenocytes. Diabetes 2002; 51:356 - 365
  • Panakanti R, Mahato RI. Bipartite vector encoding hVEGF and hIL-1Ra for ex vivo transduction into human islets. Mol Pharm 2009; 6:274 - 284
  • Panakanti R, Mahato RI. Bipartite adenoviral vector encoding hHGF and hIL-1Ra for improved human islet transplantation. Pharm Res 2009; 26:587 - 596
  • Narang AS, Sabek O, Gaber AO, Mahato RI. Co-expression of vascular endothelial growth factor and interleukin-1 receptor antagonist improves human islet survival and function 2006; 23:1970 - 1982
  • Kawamoto K, Tanemura M, Komoda H, Omori T, Fumimoto Y, Shimada K, et al. Adenoviral-mediated overexpression of membrane-bound human FasL and human decoy Fas protect pig islets against human CD8+ CTL-mediated cytotoxicity. Transplant Proc 2006; 38:3286 - 3288
  • Kawamoto K, Tanemura M, Saga A, Komoda H, Fumimoto Y, Deguchi T, et al. Adenoviral-mediated overexpression of either membrane-bound human FasL or human decoy Fas can prolong pig islet xenograft survival in a rat transplant model. Transplant Proc 2008; 40:477 - 479
  • Giannoukakis N, Mi Z, Rudert WA, Gambotto A, Trucco M, Robbins P. Prevention of beta cell dysfunction and apoptosis activation in human islets by adenoviral gene transfer of the insulin-like growth factor I. Gene Ther 2000; 7:2015 - 2022
  • Zhang YC, Molano RD, Pileggi A, Powers M, Cross J, Wasserfall C, et al. Adeno-associated virus transduction of islets with interleukin-4 results in impaired metabolic function in syngeneic marginal islet mass transplantation. Transplantation 2002; 74:1184 - 1186
  • Zhang YC, Pileggi A, Molano RD, Wasserfall C, Campbell-Thompson M, Ricordi C, et al. Systemic overexpression of interleukin-10 fails to protect allogeneic islet transplants in nonobese diabetic mice. Transplantation 2005; 80:530 - 533
  • Carter JD, Ellett JD, Chen M, Smith KM, Fialkow LB, McDuffie MJ, et al. Viral IL-10-mediated immune regulation in pancreatic islet transplantation. Mol Ther 2005; 12:360 - 368
  • Oh TK, Li MZ, Kim ST. Gene therapy for diabetes mellitus in rats by intramuscular injection of lentivirus containing insulin gene. Diabetes Res Clin Pract 2006; 71:233 - 240
  • Gallichan WS, Kafri T, Krahl T, Verma IM, Sarvetnick N. Lentivirus-mediated transduction of islet grafts with interleukin 4 results in sustained gene expression and protection from insulitis. Hum Gene Ther 1998; 18:2717 - 2726
  • Chou FC, Sytwu HK. Overexpression of thioredoxin in islets transduced by a lentiviral vector prolongs graft survival in autoimmune diabetic NOD mice. J Biomed Sci 2009; 16:71
  • Giannoukakis N, Mi Z, Gambotto A, Eramo A, Ricordi C, Trucco M, et al. Infection of intact human islets by a lentiviral vector. Gene Ther 1999; 6:1545 - 1551
  • Fenjves ES, Ochoa MS, Cechin S, Gay-Rabinstein C, Pérez-Alvarez I, Ichii H, et al. Protection of human pancreatic islets using a lentiviral vector expressing two genes: cFLIP and GFP. Cell Transplant 2008; 17:793 - 802

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.