1,136
Views
28
CrossRef citations to date
0
Altmetric
Special Focus Review

New routes for allergen immunotherapy

, , , &
Pages 1525-1533 | Received 05 Jul 2012, Accepted 23 Aug 2012, Published online: 01 Oct 2012

Abstract

IgE-mediated allergy is a highly prevalent disease in the industrialized world. Allergen-specific immunotherapy (SIT) should be the preferred treatment, as it has long lasting protective effects and can stop the progression of the disease. However, few allergic patients choose to undergo SIT, due to the long treatment time and potential allergic adverse events. Since the beneficial effects of SIT are mediated by antigen presenting cells inducing Th1, Treg and antibody responses, whereas the adverse events are caused by mast cells and basophils, the therapeutic window of SIT may be widened by targeting tissues rich in antigen presenting cells. Lymph nodes and the epidermis contain high density of dendritic cells and low numbers of mast cells and basophils. The epidermis has the added benefit of not being vascularised thereby reducing the chances of anaphylactic shock due to leakage of allergen. Hence, both these tissues represent highly promising routes for SIT and are the focus of discussion in this review.

Introduction

Allergic rhino-conjunctivitis affects more than 20% of the population in western Europe,Citation1 and represents a significant cause of illness with impact on daily activities and sleep quality.Citation2 While many patients respond effectively to symptomatic pharmacotherapy such as antihistamines and corticosteroids, a substantial proportion of patients report inadequate symptom alleviation and reduced quality of life (QoL).Citation3 Here, allergen-specific immunotherapy (SIT) offers a disease-specific causative treatment alternative by inducing tolerance to the allergen.Citation4 Hence, patients who suffer from allergies in which allergen avoidance is not possible, such as pollen or house dust mite allergy, patients with systemic reactions to hymenoptera venom and patients whose symptoms have not responded adequately to optimal pharmacotherapy are usually the prime candidates for SIT.Citation5

Until recently, SIT was predominantly administered subcutaneously (SCIT) as described in the early works by Noon and Freeman.Citation6,Citation7 SCIT comprises repeated administration of allergen extracts, of as many as 50–80 injections over a period of at least three years, but for immunotherapy to be effective, careful patient selection is required.Citation8,Citation9 Provided adequate precautions are taken, SIT is safe,Citation10 but a decision whether to treat with immunotherapy will depend on a variety of factors, and conventional SCIT has a low appeal for most patients. Only approximately 5% of patients with allergic rhino-conjunctivitis choose to undergo immunotherapy, due to the long treatment duration with severe impact on daily life, e.g., numerous doctor visits with consequential absence from school or work. Personal and organizational factors then also determine whether one type of immunotherapy is more suitable than another, e.g., SCIT or the more patient-friendly, but equally long and less validated sublingual immunotherapy (SLIT), which can be self-administered. In response to the drawbacks of SCIT and SLIT, current SIT research in allergy focuses on developing methods that are more patient-friendly in order to facilitate better patient compliance.Citation4,Citation5,Citation11-Citation13

One of the major challenges in this field of research is to balance efficacy and safety of the new SIT methods. Immunologically, SIT and conventional vaccination do not substantially differ.Citation14 In conventional childhood vaccination, three injections of antigen and adjuvant are typically sufficient to induce long-term protection against vaccine-preventable diseases caused by microbes and higher doses typically produce stronger immune responses. From a qualitative viewpoint, it would be beneficial to use vaccines that are able to shift the type of immune response from allergy-associated T-helper type 2 (Th2) toward the more protective Th1 and Treg- based immune responses. This could be achieved by using higher doses of allergen with stronger Th1-stimulating adjuvants. However, in conventional SIT, high doses of antigen cannot be used since patients are allergic to the antigen used. Therefore, antigen doses in conventional SIT must be kept low to reduce the risk of patients suffering from local side effects and systemic and serious adverse effects. Hence, the goal of current SIT research is to allow strong stimulation of the immune system, while bypassing potential adverse reactions.

Besides using hypo-allergenic and recombinant allergens, one strategy to improve the therapeutic window of SIT is to choose administration routes that allow efficient targeting of potent professional antigen presenting cells (APCs), while avoiding tissues with high density of mast cells. Moreover, SIT in highly vascularized tissues should also be avoided in order to avoid vascular leakage of allergen, which can cause systemic allergic reactions or anaphylaxis. Two tissues which fit the above-described properties are the epidermal layer of the skin and the lymph nodes; the skin being the largest and most accessible organ system and the lymph nodes, the organs with the highest density of immune competent cells as well as being the final target of any vaccine antigen or SIT allergen. During the last decade, we and others have studied the use of these routes for vaccination and immunotherapy () and in this review, we will discuss the outcomes of the research with a special focus on allergen-specific immunotherapy.

Table 1. Clinical trials with allergen-specific intralymphatic (ILIT) or epicutaneous (EPIT) immunotherapy

Epicutaneous Immunotherapy (EPIT)

The skin as an immunological organ in topical vaccination

Briefly, the human skin consists of the epidermis and the underlying dermis. Although the epidermis of the palms and soles are thicker by 1 mm, it typically forms a 50–150 μm thick mechanical barrier with a 15–20 μm thick outer layer of cornified keratinocytes in a lipid-rich matrix known as the stratum corneum, which effectively excludes the entry of large molecules.Citation15-Citation17 While predominantly consisting of keratinocytes, the epidermis also contains specialized cells such as pigment-producing melanocytes and antigen-presenting Langerhans cells (LCs).Citation16 LCs account for only 3–5% of the epidermal cells, but are attractive targets for vaccines as they cover up to 20% of the skin surface by forming a network with their dendrites.Citation15,Citation18 The non-vascularized epidermis is separated by the basement membrane from the more complex dermis, which harbors a great diversity of cells as well as lymphatic and vascular conduits. Besides fibroblasts, macrophages and mast cells, the dermis comprises different subsets of dermal dendritic cells (DCs), as well as, depending on the inflammatory stage, other immune cells of myeloid or lymphoid origin.Citation16,Citation19

The skin with its associated lymphoid tissue (SALT) is considered an immunologically competent organ system.Citation20 Although many of the immunologic properties of the skin are known,Citation21 specific functions of the different DC subsets and the keratinocytes with regard to the stimulation and shaping of adaptive immune response remains to be unravelled.Citation22,Citation23 Whereas epidermal LCs have been demonstrated to hold a key role in elicitation of CD8+ T-cell responses and in shaping Th2-type/Treg-type responses, activation of dermal DC subsets has been found to be essential for B cell class switching and induction of a Th1-type response.Citation24,Citation25 In line with this, dermal DC subsets were observed to preferentially localize to the B-cell areas of draining lymph nodes, whereas LCs were observed to migrate to the T-cell areas.Citation26 While previously considered mostly immunologically inactive and only forming part of the physical barrier function of the skin, keratinocytes have now been recognized to play a pivotal role in triggering and guiding the adaptive immune responses.Citation23,Citation27 These epithelial cells express different molecular mediators under different conditions, thereby governing dictating a variety of responses. A slight stress to the epithelium, such as abrasion, may trigger secretion of the IL-7-like cytokine thymic stromal lymphopoietin (TSLP), IL-25 and IL-33, which in turn instruct non-inflammatory Treg- or Th2-type responses.Citation28-Citation32 On the other hand, epithelial damages may trigger expression of additional molecules such as IL-1α, IL-6 and TNF-α, which skew Th1-type responses.Citation27 The degree of epithelial damage might explain the observed dichotomy of different DC subsets: while mild irritations induce non-inflammatory responses transmitted by LCs, stronger epithelial damages induce pro-inflammatory response performed by dermal DCs. The exact understanding of these processes would certainly support a more rational design of vaccines for epi- or transcutaneous administration.

Although the skin is easily accessible, topical application of a vaccine in a cream or in a patch does not typically induce an immune response because of the low permeability of vaccines through the stratum corneum. When, in ancient times, the Indians first used the skin to vaccinate against smallpox, the stratum corneum was disrupted by scratching also called scarification.Citation33 Today, this has been replaced by tape stripping and other abrasive methods,Citation34,Citation35 which aim at gently removing the cornified keratinocytes without disrupting the underlying basement membrane. Similarly, but more precisely, APCs of the epidermis can nowadays be directly targeted by using microneedle arrays with defined needle lengths thanks to latest technological progress.Citation15,Citation36-Citation42 These methods are associated with physical irritation of keratinocytes, leading to secretion of cytokines, which in turn facilitates activation of the immune system.Citation27,Citation43 Alternatively, penetration of antigens through the epidermis can be enhanced by skin hydration over a time period of at least 4 to 10 h.Citation44 This is typically achieved by application of an occlusive patch leading to sweat accumulation.Citation45,Citation46

Allergen-specific immunotherapy by topical epicutaneous administration

Despite the ancient success of epicutaneous smallpox vaccination,Citation47 this route of administration did not attract major attention until the end of the 20th century, when Glenn and coworkers demonstrated efficacious transcutaneous vaccination against numerous infectious diseases as well as cancer.Citation33,Citation34,Citation48-Citation53. A decade later, our group published the first placebo controlled trial on allergen-specific epicutaneous immunotherapy (EPIT).Citation54

However, the first records describing the administration of allergens via the skin date back to 1929. Soon after the introduction of SCIT by Leonard Noon,Citation6 the risk of suffering from anaphylaxis and other allergic adverse events upon subcutaneous allergen administration was recognized as a real problemCitation55 and spurred testing of intradermal pollen extract administrations.Citation56 Similar to the intradermal allergy testing of today, pollen extracts were applied to the skin. The authors reported that the treatment was safe and highly successful with symptom amelioration in all the 29 patients after administration of three doses.

Another event that suggested the potential suitability of EPIT was an observation reported in 1921, when an asthmatic patient experienced complete symptom relief after administration of horse dander on scarificated skin, a method called “cutiréactions répétées.”Citation57 Then, French allergologists paved the way for EPIT during the 1950s and 60s. Pautrizel and coworkersCitation58 administered allergen extracts onto epidermis that was slightly rubbed, while the team of BlamoutierCitation59,Citation60 applied allergen drops onto heavily scarified skin, a method called “méthode de quadrillage cutané.” Both groups observed high treatment success, but while the former method required a large number of applications the method of Blamoutier could be performed co-seasonally with significant symptom amelioration after an average of four treatments. Both methods were associated with fewer and milder allergic side effects than typically observed with SCIT.Citation58-Citation60 Several confirmatory trials were performed in the following years by the Swiss clinicians Eichenberger and Storck, who reported a treatment success rate of approximately 80%.Citation61 Symptoms rapidly diminished after a single application, and the effect lasted from three days to three weeks. Hence, 6 to 12 treatments were required for a symptom free season and the method was reported to be also effective in patients who responded inadequately to conventional SCIT.Citation61-Citation63

Inspired by these early reports, we performed the first randomized placebo-controlled study that tested the clinical efficacy and safety of EPIT in patients allergic to grass pollen.Citation54 Twelve patches with allergen or placebo were applied on the upper arm, before and during the pollen season, and the application area was prepared by tape stripping.Citation64-Citation67 A 70% improvement of hay fever symptoms in the treated patients was observed, while the placebo effect accounted for 20%. No severe systemic allergic reactions were observed, but local eczematous skin reactions were frequently reported.

The first study was followed up with another phase I/IIa trial with 132 patients in order to define optimal allergen doses.Citation68 Hay-fever symptoms during the pollen season were reduced by more than 30% in the first year and by 24% in the second year in the high-dose group when compared with those in the placebo group. The alleviation of symptoms in the follow-up year was dependent on the treatment dose, but higher allergen doses were associated with more local adverse events (AEs), manifested by pruritus, erythema, wheal or eczema. Eleven (8.3%) systemic AEs of grades 1 to 2 required treatment and led to study exclusion. No drug-related serious AE was recorded.

A third placebo-controlled trial, including 98 patients, was initiated in 2009 and recently completed (NCT00777374). The goal of this study was to further compare the efficacy of EPIT using the combined symptom medication score as well measuring immunological responses. Results are expected in 2012. In support of our results, Agostinis and coworkers recently demonstrated efficacy and safety of EPIT when used as a treatment for grass pollen allergy in children.Citation69 Hay fever symptoms as well as antihistamine use were significantly reduced in the active treatment group.

Furthermore the French company DBV Technologies currently develops an occlusive patch for EPIT. It is known that the atopy patch test can induce an eczematous skin reaction even on intact skin.Citation70 This suggests efficient antigen penetration, and DBV Technologies has confirmed this with an occlusive patch initially developed for diagnostic purposes (Diallertest).Citation45,Citation46,Citation71 Known commercially as Viaskin®, this epidermal delivery system (EDS) relies on the ability to deliver whole protein molecules to the skin, facilitated through hydration of the stratum corneum under an occlusive chamber. Whole protein, delivered via such a system, has been demonstrated to accumulate in the stratum corneum, from where it diffuses down to immune cells in the viable layers of the epidermis.Citation46,Citation71,Citation72 Murine studies, designed to test therapeutic efficacy of the Viaskin® EDS, demonstrated equivalence of EPIT and SCITCitation46 as well as EPIT and SLITCitation73 in preventing allergic airway reactions upon airway allergen challenge. Moreover, EPIT using the Viaskin® EDS was also efficient in the treatment of food allergy as measured by prevention of mast cell degranulation upon oral allergen challenge.Citation71,Citation74 A clinical pilot trial was launched to test efficacy and safety of Viaskin® EDS EPIT in children with allergy to cow milk. The treatment was well tolerated with no serious systemic allergic reaction, but there was a significant increase in local eczematous reactions.Citation45 A trend toward greater tolerance to cow milk was observed in the treated group upon food provocation. Phase I (NCT01170286) and phase II trials (NCT01197053) have been initiated to substantiate these findings when using Viaskin® EDS for treatment against peanut allergy.

Intralymphatic Immunotherapy

Immunological specialties of the lymph nodes

T- and B-cell receptors are randomly rearranged to generate a large repertoire of lymphocytes that can specifically recognize all possible antigens. Conversely, however, the frequencies of antigen-specific lymphocytes are so low (approximately one in ten million) that antigens must be presented to millions of T and B cells in order to find their match and to elicit a response. Therefore, antigens need to either drain into secondary lymphatic organs or be transported there. Here, antigens can be presented to high numbers of T and B cells for stimulation of an immune response.Citation75 Whereas antigens staying outside of secondary lymphoid organs have little chance to encounter specific T or B cells and are therefore poorly immunogenicCitation76-Citation78.

Lymph vessels drain substances from the interstitial fluid into regional lymph nodes with highly variable efficiency. As lymph vessels have evolved to drain pathogens into lymph nodes, viruses and small particles (20–200 nm) drain freely and efficiently from the peripheral sites into lymph nodes. However, only a low percentage of the particles drain into the lymph nodes.Citation79 Larger bacteria and particles (500–2000 nm) are mostly transported into lymph nodes by DCs and other APCs. On the other hand, drainage of non-particulate antigens from the periphery to lymph nodes is typically less efficient and only one thousandth to one millionth of the antigen dose reaches the lymph nodes. Because many of the currently used vaccines are non-particulate, one may expect that the direct administration of a vaccine into a lymph node would increase antigen presentation and stimulation of immune responses.

Intralymphatic immunotherapy

A large number of studies, both pre-clinical and clinical have been based on the intralymphatic administration of drug or vaccines. In humans, the drug or vaccine preparations are typically administered intralymphatically into an inguinal lymph node under ultrasound control, a procedure that is uncomplicated and performed routinely by sonologists as well as by radiologists for the administration of imaging agents. For a comprehensive review, see references Citation12 and Citation80Citation83.

Antigen pulsed DCs injected into the lymph node localize to the paracortex.Citation84-Citation86 As DCs and T cells are present at very high densities in lymph nodes, co-stimulatory signals for T- and B-cell induction may be provided as a bystander effect. While some clinical trials using intranodal therapy with DCs have suggested enhanced immune responses,Citation87,Citation88other trials failed to demonstrate advantage of intranodal over intradermal DCs delivery.Citation84,Citation89 We also found that non-professional APCs, such as a fibro-sarcoma cell line efficiently induced antigen specific CD8+ T-cell responses in lymph nodes via direct antigen presentation on the MHC class I molecule present on the fibro-sarcoma.Citation76,Citation90 Intranodal therapy with tumor cells has been tried in both human cancer patients and dogs with indication of success.Citation91-Citation95

Direct administration of MHC class I binding peptide vaccines into lymph nodes or the spleen has been demonstrated to strongly enhance CD8 T-cell responses that were protective against viral challenge and tumor growth in mice.Citation96 Similarly, intranodal immunisation also dramatically enhanced the efficacy of plasmid DNA vaccination in miceCitation97-Citation99 as well as RNA vaccination.Citation100-Citation102

Intranodal immunisation with proteins for induction of antibodies was performed surprisingly early. At a time when it was difficult to purify high quantities of proteins, researchers were looking for a more efficient route of immunisation.Citation103 By intralymphatic immunotherapy, only nanograms of proteins were required to elicit sufficiently strong immune responses.Citation104 Likewise, targeted lymph node administration is also extensively documented to be the most efficient way to immunize macaques against SIV.Citation105 Similar results were obtained in macaques vaccinated with other proteins.Citation106-Citation111

Several clinical trials with intranodal therapy have confirmed these initial pre-clinical studies. Most of these studies have been performed in cancer patients usually as autologous vaccination using antigen-pulsed DCs.Citation88,Citation112-Citation118 This treatment was typically safe and stimulated antigen-specific and cytotoxic CD8 T-cell responses in the patients. Although intralymphatic immunotherapy with DCs has been associated with improved survival,Citation112 increased immune responses were not always followed by marked clinical benefits.Citation115 In other cancer trials, the intranodal vaccines were based on plasmid DNACitation119 or plasmid DNA and peptide in a prime-boost regime.Citation120,Citation121 Finally, intranodal injections of vaccines based on viral vectors have been studied in melanoma patients, who responded with strong cytotoxic and other immunological T-cell responses as well as some clinical benefits.Citation122,Citation123

Allergen-specific intralymphatic immunotherapy (ILIT)

As mentioned above, IgE-mediated allergies, such as allergic rhino-conjunctivitis and asthma, have become highly prevalentCitation124-Citation127 and the gold standard of therapy is subcutaneous allergen-specific immunotherapy (SCIT), which confers long-term benefits, and which interrupts with the atopic march that is the expansion of allergic sensitizations to other allergensCitation128 and the progression from allergic rhinitis to asthma.Citation129 From a medical perspective, SCIT is therefore superior to symptomatic treatments, but its efficacy should be improved and the treatment time reduced to make SIT more patient friendly.

Therefore, we have investigated whether SCIT could be enhanced by intralymphatic administration of allergens. In mice, we have demonstrated that ILIT with a variety of allergens, e.g., bee venom phospholipase-A2, ovalbumin and allergen extracts from grass pollen, birch pollen and cat dander, stimulated anti-allergic and protective B- and T-cell immune responses.Citation130-Citation134 In fact, changing from the subcutaneous route to direct intralymphatic injection significantly enhanced the efficacy of immunization, inducing allergen-specific IgG2a antibody responses that were 10–20 times higher with only 0.1% of the allergen dose. Since allergic side effects are proportional to the allergen dose, intralymphatic SIT should also have the potential to reduce side effects, since lower doses of allergens are required for efficacious SIT. Moreover, allergen ILIT enhanced IL-1, IFN-γ, IL-4 and IL-10 secretion when compared with subcutaneous SIT, suggesting that ILIT did not polarize the immune response allergen toward Th1, Th2 or Tregs, but generated overall stronger responses.

Studies of biodistribution in mice showed that approximately 100-fold higher allergen doses reached the lymph nodes after intralymphatic than after subcutaneous injection in the same draining region.Citation132 Similar results were obtained in humans when intralymphatic injections were compared with subcutaneous injection using radio tracing.Citation81,Citation83 Essentially, when the same dose of a 99mTc labeled protein was injected into either a superficial inguinal lymph node or subcutaneously at a site just 10 cm above the contralateral inguinal lymph node, only a small fraction of the subcutaneously administered protein reached the lymph nodes after 4 h and this fraction did not increase after 24 h. However, within 20 min of the intralymphatic injection, the protein drained into the deep subcutaneous lymph nodes and even further into a pelvic lymph node. These experiments clearly demonstrated the potential of allergen ILIT to that of allergen SCIT.

Clinical trials with allergen ILIT

In a first clinical trial, eight patients with bee-venom allergy grade III, who would have received approximately 70 subcutaneous injections of each 100 µg bee venom extract under normal SCIT protocol, received three injections of only 10 µg directly into the inguinal lymph node. In this proof of concept trial, seven out of the eight treated patients were protected against a subsequent bee sting challenge. In a larger multicenter clinical trial conducted after the pilot study with 66 grade III and IV bee venom allergic patients, allergen-ILIT mediated protection was shown to correlate with immunological parameters (Senti et al., manuscript in preparation). The safety and efficacy of intranodal bee venom allergen extract (Bee AlleVax) was further tested in a multicenter study. Between 2001 and 2003, 67 subjects from 15 centers across Europe and in Australia were randomized to receive either 10 or 20 µg doses of Bee AlleVax via intranodal injections. Clinical endpoints included bee venom specific IgE and IgG serum levels and the degree of protection after a bee sting challenge. Desensitization by ILIT not only resulted in increased IgG and IgE levels, but also in a high number of serious adverse events occurring both after ILIT and after bee sting challenge. For this reason, the study was terminated by the sponsors. The high number of serious adverse events may suggest that the immunological protection offered by Bee AlleVax was insufficient. Two confounding factors may explain the failure of Bee AlleVax. First, a major fraction of the bee venom extract remained un-absorbed to the alum (personal communication from S.J. McCormack, former CEO of AlleCure Corp.). This may cause vascular leakage of free allergen. Second, the bee venom allergen extract was prepared in US using American honey bees and the studies were done on patients from Europe and Australia, and it is known that continental differences in the content of bee venom allergens exists.

In another Swiss trial, 165 patients with grass pollen-induced rhino-conjunctivitis were randomized to receive either 54 subcutaneous injections with pollen extract over three years [cumulative allergen dose 4,031,540 standardized quality units (SQ-U)] or three intralymphatic injections over two months (cumulative allergen dose 3,000 SQ-U); the trial was monocentric and open labeled.Citation135 Patients were evaluated after four months, one year and three years. Three low-dose intralymphatic allergen administrations increased tolerance upon nasal provocation with pollen within four months after treatment. Tolerance was long lasting and equivalent to that achieved after standard SCIT. Allergen ILIT ameliorated hay fever symptoms, reduced skin prick test reactivity, decreased specific IgE in serum and caused fewer adverse events than did SCIT. ILIT also enhanced compliance and was less painful than venous puncture. In conclusion, intralymphatic allergen administration enhanced safety and efficacy of immunotherapy and could reduce treatment time from three years to eight weeks.

In a recently reported Danish clinical trial on ILIT in grass pollen allergic patients by Malling and coworkers, three and six low dose intralymphatic injections were found to induce IgG4, but in contrast to the described Swiss trial no reduction in hay fever symptoms could be found.Citation136 Hay fever symptoms actually worsened when compared with placebo, and IgE was boosted after allergen ILIT. In the Danish trial, intralymphatic grass pollen extract injections were given on a weekly interval, and not monthly, as in the Swiss trial.Citation135 Experience from vaccine immunology recommends a time interval between priming injections of at least three weeks to allow development of successive waves of antigen specific primary responses and affinity maturation of memory B cells.Citation137 Also, the continued antigen presentation generated by weekly administration in the Danish trial by Malling and coworkers may have favored boosting of Th2 responses, as has been previously demonstrated in mouse models.Citation138 With the on-and-off allergen profile generated by monthly injections in the Swiss trial, no boosting of IgE was observed.Citation135

Recently, we applied ILIT using a MHC class II-targeting cat dander allergen; a recombinant Fel d 1 conjugated to a TAT translocation sequence and human invariant chain (MAT-Fel d 1). In this randomized, placebo-controlled and double-blind trial, ILIT with MAT-Fel d 1 in alum was compared with ILIT with placebo in cat dander allergic patients.Citation139 Three monthly injections with MAT-Fel d 1 elicited no adverse events, and there was significant increase in allergen tolerance after nasal provocation. In addition, allergen ILIT stimulated regulatory T-cell responses with IL-10 cytokine secretion and increased cat dander-specific IgG4 production.

Conclusions

The ideal route for allergen-specific immunotherapy is one with a high density of potent professional APCs, so that immunogenicity of the allergen is increased enabling dose-reduction as well as reduction of the number of allergen administrations. Also the ideal route of allergen immunotherapy should contain only few mast cells to avoid local allergic reactions, and vascularization should be poor to minimize the risk of allergen leakage or inadvertent intravascular allergen administration entailing severe anaphylactic side effects. Both, the epidermis and the lymph node contain a high density of DCs. While lymph nodes contain very few mast cells, the epidermis contains none. Further advantages of the epidermis as route of allergen administration are the accessibility and fact that the epithelium is not vascularized.

It has been verified in clinical trials that both safety and efficacy of intranodal SIT (ILIT) is improved when compared with conventional subcutaneous SIT (SCIT). For epicutaneous SIT (EPIT), clinical comparisons with SCIT have not yet been donebecause current guidelines for testing efficacy of SIT require placebo control instead of SCIT control. The type of allergens suitable for ILIT and EPIT are in principle the same as those suitable for SCIT. However, from a safety viewpoint, SIT with allergens that have very high affinity to IgE would surely benefit from ILIT as ILIT enables effective therapy with much lower doses than those required in effective SCIT. In conclusion, both the lymph node and the epidermis represent highly interesting routes for allergen immunotherapy, combining the advantages of high density of APCs and low frequency of mast cells and vascular structures, factors that enable dose reduction, reduction of doctor visits and side effects.

Abbreviations:
SIT=

allergen specific immunotherapy

ILIT=

intralymphatic immunotherapy

EPIT=

epicutaneous immunotherapy

SLIT=

sublingual immunotherapy

QoL=

quality of life

DC=

dendritic cells

LCs=

Langerhans cells

Antigen presenting cells=

APCs

Disclosure of Potential Conflicts of Interest

T.M.K. and G.S. are named as inventors on a patent on epicutaneous immunotherapy, and T.M.K. is named as the inventor on a patent on intralymphatic immunotherapy. Both patents are owned by the University of Zurich. P.J., S.M. and D.M. declare to have no relevant conflicts of interest.

References

  • Bauchau V, Durham SR. Prevalence and rate of diagnosis of allergic rhinitis in Europe. Eur Respir J 2004; 24:758 - 64; http://dx.doi.org/10.1183/09031936.04.00013904; PMID: 15516669
  • Scadding GK, Durham SR, Mirakian R, Jones NS, Leech SC, Farooque S, et al, British Society for Allergy and Clinical Immunology. BSACI guidelines for the management of allergic and non-allergic rhinitis. Clin Exp Allergy 2008; 38:19 - 42; http://dx.doi.org/10.1111/j.1365-2222.2007.02888.x; PMID: 18081563
  • Smith H. Asthma, inflammation, eosinophils and bronchial hyperresponsiveness. Clin Exp Allergy 1992; 22:187 - 97; http://dx.doi.org/10.1111/j.1365-2222.1992.tb03072.x; PMID: 1571812
  • Frew AJ. Allergen immunotherapy. J Allergy Clin Immunol 2010; 125:Suppl 2 S306 - 13; http://dx.doi.org/10.1016/j.jaci.2009.10.064; PMID: 20176266
  • Walker SM, Durham SR, Till SJ, Roberts G, Corrigan CJ, Leech SC, et al, British Society for Allergy and Clinical Immunology. Immunotherapy for allergic rhinitis. Clin Exp Allergy 2011; 41:1177 - 200; http://dx.doi.org/10.1111/j.1365-2222.2011.03794.x; PMID: 21848757
  • Noon L. Prophylactic inoculations against hay fever. Lancet 1911; 177:1572 - 3; http://dx.doi.org/10.1016/S0140-6736(00)78276-6
  • Freeman J. Further observations on the treatment of hay fever by hypodermic inoculations of pollen vaccine. Lancet 1911; 178:814 - 7; http://dx.doi.org/10.1016/S0140-6736(01)40417-X
  • Alvarez-Cuesta E, Bousquet J, Canonica GW, Durham SR, Malling HJ, Valovirta E, EAACI, Immunotherapy Task Force. Standards for practical allergen-specific immunotherapy. Allergy 2006; 61:Suppl 82 1 - 20; http://dx.doi.org/10.1111/j.1398-9995.2006.01219_1.x; PMID: 16930249
  • Calderón MA, Casale TB, Togias A, Bousquet J, Durham SR, Demoly P. Allergen-specific immunotherapy for respiratory allergies: from meta-analysis to registration and beyond. J Allergy Clin Immunol 2011; 127:30 - 8; http://dx.doi.org/10.1016/j.jaci.2010.08.024; PMID: 20965551
  • Calderon MA, Alves B, Jacobson M, Hurwitz B, Sheikh A, Durham S. Allergen injection immunotherapy for seasonal allergic rhinitis. Cochrane Database Syst Rev 2007; CD001936; PMID: 17253469
  • Nelson HS. Allergen immunotherapy: where is it now?. J Allergy Clin Immunol 2007; 119:769 - 79; http://dx.doi.org/10.1016/j.jaci.2007.01.036; PMID: 17337297
  • von Moos S, Kündig TM, Senti G. Novel administration routes for allergen-specific immunotherapy: a review of intralymphatic and epicutaneous allergen-specific immunotherapy. [xi.] Immunol Allergy Clin North Am 2011; 31:391 - 406, xi; http://dx.doi.org/10.1016/j.iac.2011.02.012; PMID: 21530827
  • Senti G, Martínez Gómez JM, Rettig L, Wüthrich B, Kündig TM, Johansen P. Immunotherapeutic targeting of allergic disease. Inflamm Allergy Drug Targets 2006; 5:243 - 52; http://dx.doi.org/10.2174/187152806779010963; PMID: 17168795
  • Kündig TM, Bachmann MF. Allergen-specific immunotherapy: regulatory T cells or allergen-specific IgG?. Hum Vaccin 2010; 6:673 - 5; http://dx.doi.org/10.4161/hv.6.8.12007; PMID: 20523122
  • Bal SM, Ding Z, van Riet E, Jiskoot W, Bouwstra JA. Advances in transcutaneous vaccine delivery: do all ways lead to Rome?. J Control Release 2010; 148:266 - 82; http://dx.doi.org/10.1016/j.jconrel.2010.09.018; PMID: 20869998
  • Nestle FO, Di Meglio P, Qin JZ, Nickoloff BJ. Skin immune sentinels in health and disease. Nat Rev Immunol 2009; 9:679 - 91; PMID: 19763149
  • Bos JD, Meinardi MM. The 500 Dalton rule for the skin penetration of chemical compounds and drugs. Exp Dermatol 2000; 9:165 - 9; http://dx.doi.org/10.1034/j.1600-0625.2000.009003165.x; PMID: 10839713
  • Babiuk S, Baca-Estrada M, Babiuk LA, Ewen C, Foldvari M. Cutaneous vaccination: the skin as an immunologically active tissue and the challenge of antigen delivery. J Control Release 2000; 66:199 - 214; http://dx.doi.org/10.1016/S0168-3659(99)00274-6; PMID: 10742580
  • Merad M, Ginhoux F, Collin M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol 2008; 8:935 - 47; http://dx.doi.org/10.1038/nri2455; PMID: 19029989
  • Streilein JW. Skin-associated lymphoid tissues (SALT): origins and functions. J Invest Dermatol 1983; 80:Suppl 12s - 6s; http://dx.doi.org/10.1111/1523-1747.ep12536743; PMID: 6602189
  • Kupper TS, Fuhlbrigge RC. Immune surveillance in the skin: mechanisms and clinical consequences. Nat Rev Immunol 2004; 4:211 - 22; http://dx.doi.org/10.1038/nri1310; PMID: 15039758
  • Kapsenberg ML. Dendritic-cell control of pathogen-driven T-cell polarization. Nat Rev Immunol 2003; 3:984 - 93; http://dx.doi.org/10.1038/nri1246; PMID: 14647480
  • Matzinger P. Friendly and dangerous signals: is the tissue in control?. Nat Immunol 2007; 8:11 - 3; http://dx.doi.org/10.1038/ni0107-11; PMID: 17179963
  • Klechevsky E, Morita R, Liu M, Cao Y, Coquery S, Thompson-Snipes L, et al. Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity 2008; 29:497 - 510; http://dx.doi.org/10.1016/j.immuni.2008.07.013; PMID: 18789730
  • Ueno H, Schmitt N, Klechevsky E, Pedroza-Gonzalez A, Matsui T, Zurawski G, et al. Harnessing human dendritic cell subsets for medicine. Immunol Rev 2010; 234:199 - 212; http://dx.doi.org/10.1111/j.0105-2896.2009.00884.x; PMID: 20193020
  • Kissenpfennig A, Henri S, Dubois B, Laplace-Builhé C, Perrin P, Romani N, et al. Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity 2005; 22:643 - 54; http://dx.doi.org/10.1016/j.immuni.2005.04.004; PMID: 15894281
  • Swamy M, Jamora C, Havran W, Hayday A. Epithelial decision makers: in search of the ‘epimmunome’. Nat Immunol 2010; 11:656 - 65; http://dx.doi.org/10.1038/ni.1905; PMID: 20644571
  • Cevikbas F, Steinhoff M. IL-33: a novel danger signal system in atopic dermatitis. J Invest Dermatol 2012; 132:1326 - 9; http://dx.doi.org/10.1038/jid.2012.66; PMID: 22499037
  • Gilliet M, Soumelis V, Watanabe N, Hanabuchi S, Antonenko S, de Waal-Malefyt R, et al. Human dendritic cells activated by TSLP and CD40L induce proallergic cytotoxic T cells. J Exp Med 2003; 197:1059 - 63; http://dx.doi.org/10.1084/jem.20030240; PMID: 12707303
  • Savinko T, Matikainen S, Saarialho-Kere U, Lehto M, Wang G, Lehtimäki S, et al. IL-33 and ST2 in atopic dermatitis: expression profiles and modulation by triggering factors. J Invest Dermatol 2012; 132:1392 - 400; http://dx.doi.org/10.1038/jid.2011.446; PMID: 22277940
  • Soumelis V. TSLP: from allergy to vaccine adjuvant. Eur J Immunol 2012; 42:293 - 5; http://dx.doi.org/10.1002/eji.201142337; PMID: 22266716
  • Soumelis V, Reche PA, Kanzler H, Yuan W, Edward G, Homey B, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol 2002; 3:673 - 80; PMID: 12055625
  • Mitragotri S. Immunization without needles. Nat Rev Immunol 2005; 5:905 - 16; http://dx.doi.org/10.1038/nri1728; PMID: 16239901
  • Frech SA, Dupont HL, Bourgeois AL, McKenzie R, Belkind-Gerson J, Figueroa JF, et al. Use of a patch containing heat-labile toxin from Escherichia coli against travellers’ diarrhoea: a phase II, randomised, double-blind, placebo-controlled field trial. Lancet 2008; 371:2019 - 25; http://dx.doi.org/10.1016/S0140-6736(08)60839-9; PMID: 18554712
  • Frerichs DM, Ellingsworth LR, Frech SA, Flyer DC, Villar CP, Yu J, et al. Controlled, single-step, stratum corneum disruption as a pretreatment for immunization via a patch. Vaccine 2008; 26:2782 - 7; http://dx.doi.org/10.1016/j.vaccine.2008.02.070; PMID: 18455283
  • Sullivan SP, Koutsonanos DG, Del Pilar Martin M, Lee JW, Zarnitsyn V, Choi SO, et al. Dissolving polymer microneedle patches for influenza vaccination. Nat Med 2010; 16:915 - 20; http://dx.doi.org/10.1038/nm.2182; PMID: 20639891
  • Enfield J, O’Connell ML, Lawlor K, Jonathan E, O’Mahony C, Leahy M. In-vivo dynamic characterization of microneedle skin penetration using optical coherence tomography. J Biomed Opt 2010; 15:046001; http://dx.doi.org/10.1117/1.3463002; PMID: 20799803
  • Fernando GJ, Chen X, Prow TW, Crichton ML, Fairmaid EJ, Roberts MS, et al. Potent immunity to low doses of influenza vaccine by probabilistic guided micro-targeted skin delivery in a mouse model. PLoS ONE 2010; 5:e10266; http://dx.doi.org/10.1371/journal.pone.0010266; PMID: 20422002
  • Haq MI, Smith E, John DN, Kalavala M, Edwards C, Anstey A, et al. Clinical administration of microneedles: skin puncture, pain and sensation. Biomed Microdevices 2009; 11:35 - 47; http://dx.doi.org/10.1007/s10544-008-9208-1; PMID: 18663579
  • Kim YC, Jarrahian C, Zehrung D, Mitragotri S, Prausnitz MR. Delivery systems for intradermal vaccination. Curr Top Microbiol Immunol 2012; 351:77 - 112; http://dx.doi.org/10.1007/82_2011_123; PMID: 21472533
  • van der Maaden K, Jiskoot W, Bouwstra J. Microneedle technologies for (trans)dermal drug and vaccine delivery. J Control Release 2012; 161:645 - 55; http://dx.doi.org/10.1016/j.jconrel.2012.01.042; PMID: 22342643
  • Vrdoljak A, McGrath MG, Carey JB, Draper SJ, Hill AV, O’Mahony C, et al. Coated microneedle arrays for transcutaneous delivery of live virus vaccines. J Control Release 2012; 159:34 - 42; http://dx.doi.org/10.1016/j.jconrel.2011.12.026; PMID: 22245683
  • Wood LC, Jackson SM, Elias PM, Grunfeld C, Feingold KR. Cutaneous barrier perturbation stimulates cytokine production in the epidermis of mice. J Clin Invest 1992; 90:482 - 7; http://dx.doi.org/10.1172/JCI115884; PMID: 1644919
  • Tan G, Xu P, Lawson LB, He J, Freytag LC, Clements JD, et al. Hydration effects on skin microstructure as probed by high-resolution cryo-scanning electron microscopy and mechanistic implications to enhanced transcutaneous delivery of biomacromolecules. J Pharm Sci 2010; 99:730 - 40; PMID: 19582754
  • Dupont C, Kalach N, Soulaines P, Legoué-Morillon S, Piloquet H, Benhamou PH. Cow’s milk epicutaneous immunotherapy in children: a pilot trial of safety, acceptability, and impact on allergic reactivity. J Allergy Clin Immunol 2010; 125:1165 - 7; http://dx.doi.org/10.1016/j.jaci.2010.02.029; PMID: 20451043
  • Mondoulet L, Dioszeghy V, Ligouis M, Dhelft V, Dupont C, Benhamou PH. Epicutaneous immunotherapy on intact skin using a new delivery system in a murine model of allergy. Clin Exp Allergy 2010; 40:659 - 67; http://dx.doi.org/10.1111/j.1365-2222.2009.03430.x; PMID: 20002446
  • Eyler JM. Smallpox in history: the birth, death, and impact of a dread disease. J Lab Clin Med 2003; 142:216 - 20; http://dx.doi.org/10.1016/S0022-2143(03)00102-1; PMID: 14625526
  • Jodar L, Duclos P, Milstien JB, Griffiths E, Aguado MT, Clements CJ. Ensuring vaccine safety in immunization programmes--a WHO perspective. Vaccine 2001; 19:1594 - 605; http://dx.doi.org/10.1016/S0264-410X(00)00358-3; PMID: 11166881
  • Glenn GM, Kenney RT, Ellingsworth LR, Frech SA, Hammond SA, Zoeteweij JP. Transcutaneous immunization and immunostimulant strategies: capitalizing on the immunocompetence of the skin. Expert Rev Vaccines 2003; 2:253 - 67; http://dx.doi.org/10.1586/14760584.2.2.253; PMID: 12899576
  • Glenn GM, Rao M, Matyas GR, Alving CR. Skin immunization made possible by cholera toxin. Nature 1998; 391:851; http://dx.doi.org/10.1038/36014; PMID: 9495336
  • Glenn GM, Scharton-Kersten T, Vassell R, Mallett CP, Hale TL, Alving CR. Transcutaneous immunization with cholera toxin protects mice against lethal mucosal toxin challenge. J Immunol 1998; 161:3211 - 4; PMID: 9759833
  • Glenn GM, Taylor DN, Li X, Frankel S, Montemarano A, Alving CR. Transcutaneous immunization: a human vaccine delivery strategy using a patch. Nat Med 2000; 6:1403 - 6; http://dx.doi.org/10.1038/82225; PMID: 11100128
  • Frech SA, Kenney RT, Spyr CA, Lazar H, Viret JF, Herzog C, et al. Improved immune responses to influenza vaccination in the elderly using an immunostimulant patch. Vaccine 2005; 23:946 - 50; http://dx.doi.org/10.1016/j.vaccine.2004.06.036; PMID: 15603897
  • Senti G, Graf N, Haug S, Rüedi N, von Moos S, Sonderegger T, et al. Epicutaneous allergen administration as a novel method of allergen-specific immunotherapy. J Allergy Clin Immunol 2009; 124:997 - 1002; http://dx.doi.org/10.1016/j.jaci.2009.07.019; PMID: 19733905
  • Hurwitz SH. Medicine: Seasonal Hay Fever-Some Problems in Treatment. Cal West Med 1930; 33:520 - 1; PMID: 18741454
  • Phillips EW. Relief of hay-fever by intradermal injections of pollen extract. JAMA 1926; 86:182 - 4; http://dx.doi.org/10.1001/jama.1926.02670290022008
  • Vallery-Radot P, Hangenau J. Asthme d'origine équine. Essai de désensibilisation par des cutiréactions répétées. Bull Soc Méd Hôp Paris 1921; 45:1251 - 60
  • Pautrizel R, Cabanieu G, Bricaud H, Broustet P. [Allergenic group specificity & therapeutic consequences in asthma; specific desensitization method by epicutaneous route]. Sem Hop 1957; 33:1394 - 403; PMID: 13432894
  • Blamoutier P, Blamoutier J, Guibert L. [Treatment of pollinosis with pollen extracts by the method of cutaneous quadrille ruling]. Presse Med 1959; 67:2299 - 301; PMID: 13801278
  • Blamoutier P, Blamoutier J, Guibert L. Traitement co-saisonnier de la pollinose par l'application d'extraits de pollens sur des quadrillages cutanés. Revue Francaise d'Allergie 1961; 1:112 - 20; http://dx.doi.org/10.1016/S0370-4688(61)80056-2
  • Eichenberger H, Storck H. Co-seasonal desensitization of pollinosis with the scarification-method of Blamoutier. Acta Allergol 1966; 21:261 - 7; PMID: 5953419
  • Martin-DuPan R, Buser F, Neyroud M. [Treatment of pollen allergy using the cutaneous checker square method of Blamoutier and Guibert]. Schweiz Rundsch Med Prax 1971; 60:1469 - 72; PMID: 5141542
  • Palma-Carlos A-G. Traitement co-saisonnier des pollinoses au Portugal par la méthode des quadrillages cutanés. Revue Francaise d'Allergie 1967; 7:92 - 5; http://dx.doi.org/10.1016/S0370-4688(67)80152-2
  • Dickel H, Gambichler T, Kamphowe J, Altmeyer P, Skrygan M. Standardized tape stripping prior to patch testing induces upregulation of Hsp90, Hsp70, IL-33, TNF-alpha and IL-8/CXCL8 mRNA: new insights into the involvement of 'alarmins'. Contact Dermat 2010; 63:215 - 22; http://dx.doi.org/10.1111/j.1600-0536.2010.01769.x
  • Dickel H, Goulioumis A, Gambichler T, Fluhr JW, Kamphowe J, Altmeyer P, et al. Standardized tape stripping: a practical and reproducible protocol to uniformly reduce the stratum corneum. Skin Pharmacol Physiol 2010; 23:259 - 65; http://dx.doi.org/10.1159/000314700; PMID: 20484967
  • Dickel H, Kreft B, Kuss O, Worm M, Soost S, Brasch J, et al. Increased sensitivity of patch testing by standardized tape stripping beforehand: a multicentre diagnostic accuracy study. Contact Dermatitis 2010; 62:294 - 302; http://dx.doi.org/10.1111/j.1600-0536.2010.01710.x; PMID: 20536477
  • Nickoloff BJ, Naidu Y. Perturbation of epidermal barrier function correlates with initiation of cytokine cascade in human skin. J Am Acad Dermatol 1994; 30:535 - 46; http://dx.doi.org/10.1016/S0190-9622(94)70059-1; PMID: 7512582
  • Senti G, von Moos S, Tay F, Graf N, Sonderegger T, Johansen P, et al. Epicutaneous allergen-specific immunotherapy ameliorates grass pollen-induced rhinoconjunctivitis: A double-blind, placebo-controlled dose escalation study. J Allergy Clin Immunol 2012; 129:128 - 35; http://dx.doi.org/10.1016/j.jaci.2011.08.036; PMID: 21996342
  • Agostinis F, Forti S, Di Berardino F. Grass transcutaneous immunotherapy in children with seasonal rhinoconjunctivitis. Allergy 2010; 65:410 - 1; http://dx.doi.org/10.1111/j.1398-9995.2009.02189.x; PMID: 19804450
  • Turjanmaa K, Darsow U, Niggemann B, Rancé F, Vanto T, Werfel T. EAACI/GA2LEN position paper: present status of the atopy patch test. Allergy 2006; 61:1377 - 84; http://dx.doi.org/10.1111/j.1398-9995.2006.01136.x; PMID: 17073865
  • Mondoulet L, Dioszeghy V, Vanoirbeek JA, Nemery B, Dupont C, Benhamou PH. Epicutaneous immunotherapy using a new epicutaneous delivery system in mice sensitized to peanuts. Int Arch Allergy Immunol 2011; 154:299 - 309; http://dx.doi.org/10.1159/000321822; PMID: 20962535
  • Soury D, Barratt G, Ah-Leung S, Legrand P, Chacun H, Ponchel G. Skin localization of cow’s milk proteins delivered by a new ready-to-use atopy patch test. Pharm Res 2005; 22:1530 - 6; http://dx.doi.org/10.1007/s11095-005-5881-4; PMID: 16132366
  • Mondoulet L, Dioszeghy V, Ligouis M, Dhelft V, Puteaux E, Dupont C, et al. Epicutaneous immunotherapy compared with sublingual Immunotherapy in mice sensitized to pollen (Phleum pratense). ISRN Allergy 2012; 2012:e375735; http://dx.doi.org/10.5402/2012/375735
  • Mondoulet L, Dioszeghy V, Larcher T, Ligouis M, Dhelft V, Puteaux E, et al. Epicutaneous immunotherapy (EPIT) blocks the allergic esophago-gastro-enteropathy induced by sustained oral exposure to peanuts in sensitized mice. PLoS ONE 2012; 7:e31967; http://dx.doi.org/10.1371/journal.pone.0031967; PMID: 22363776
  • Frey JR, Wenk P. Experimental studies on the pathogenesis of contact eczema in the guinea-pig. Int Arch Allergy Appl Immunol 1957; 11:81 - 100; http://dx.doi.org/10.1159/000228405; PMID: 13513182
  • Kündig TM, Bachmann MF, DiPaolo C, Simard JJ, Battegay M, Lother H, et al. Fibroblasts as efficient antigen-presenting cells in lymphoid organs. Science 1995; 268:1343 - 7; http://dx.doi.org/10.1126/science.7761853; PMID: 7761853
  • Zinkernagel RM. Localization dose and time of antigens determine immune reactivity. Semin Immunol 2000; 12:163 - 71, discussion 257-344; http://dx.doi.org/10.1006/smim.2000.0253; PMID: 10910735
  • Zinkernagel RM, Ehl S, Aichele P, Oehen S, Kündig T, Hengartner H. Antigen localisation regulates immune responses in a dose- and time-dependent fashion: a geographical view of immune reactivity. Immunol Rev 1997; 156:199 - 209; http://dx.doi.org/10.1111/j.1600-065X.1997.tb00969.x; PMID: 9176709
  • Manolova V, Flace A, Bauer M, Schwarz K, Saudan P, Bachmann MF. Nanoparticles target distinct dendritic cell populations according to their size. Eur J Immunol 2008; 38:1404 - 13; http://dx.doi.org/10.1002/eji.200737984; PMID: 18389478
  • Johansen P, Mohanan D, Martínez-Gómez JM, Kündig TM, Gander B. Lympho-geographical concepts in vaccine delivery. J Control Release 2010; 148:56 - 62; http://dx.doi.org/10.1016/j.jconrel.2010.05.019; PMID: 20562028
  • Kündig TM, Johansen P, Senti G. Intralymphatic vaccination. In: Rapuolli R, Bagnoli F, eds. Vaccine design. Norfolk (UK): Caister Academic Press, 2011:211-24.
  • Senti G, Johansen P, Kündig TM. Intralymphatic immunotherapy. Curr Opin Allergy Clin Immunol 2009; 9:537 - 43; http://dx.doi.org/10.1097/ACI.0b013e3283310ff7; PMID: 19680119
  • Senti G, Johansen P, Kündig TM. Intralymphatic immunotherapy: from the rationale to human applications. Curr Top Microbiol Immunol 2011; 352:71 - 84; http://dx.doi.org/10.1007/82_2011_133; PMID: 21725898
  • Brown K, Gao W, Alber S, Trichel A, Murphey-Corb M, Watkins SC, et al. Adenovirus-transduced dendritic cells injected into skin or lymph node prime potent simian immunodeficiency virus-specific T cell immunity in monkeys. J Immunol 2003; 171:6875 - 82; PMID: 14662894
  • De Vries IJ, Krooshoop DJ, Scharenborg NM, Lesterhuis WJ, Diepstra JH, Van Muijen GN, et al. Effective migration of antigen-pulsed dendritic cells to lymph nodes in melanoma patients is determined by their maturation state. Cancer Res 2003; 63:12 - 7; PMID: 12517769
  • de Vries IJ, Lesterhuis WJ, Barentsz JO, Verdijk P, van Krieken JH, Boerman OC, et al. Magnetic resonance tracking of dendritic cells in melanoma patients for monitoring of cellular therapy. Nat Biotechnol 2005; 23:1407 - 13; http://dx.doi.org/10.1038/nbt1154; PMID: 16258544
  • Bedrosian I, Mick R, Xu S, Nisenbaum H, Faries M, Zhang P, et al. Intranodal administration of peptide-pulsed mature dendritic cell vaccines results in superior CD8+ T-cell function in melanoma patients. J Clin Oncol 2003; 21:3826 - 35; http://dx.doi.org/10.1200/JCO.2003.04.042; PMID: 14551301
  • Lesimple T, Moisan A, Carsin A, Ollivier I, Mousseau M, Meunier B, et al. Injection by various routes of melanoma antigen-associated macrophages: biodistribution and clinical effects. Cancer Immunol Immunother 2003; 52:438 - 44; http://dx.doi.org/10.1007/s00262-003-0390-y; PMID: 12690521
  • Fong L, Brockstedt D, Benike C, Wu L, Engleman EG. Dendritic cells injected via different routes induce immunity in cancer patients. J Immunol 2001; 166:4254 - 9; PMID: 11238679
  • Ochsenbein AF, Sierro S, Odermatt B, Pericin M, Karrer U, Hermans J, et al. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature 2001; 411:1058 - 64; http://dx.doi.org/10.1038/35082583; PMID: 11429607
  • Juillard GJ, Boyer PJ. Intralymphatic immunization: current status. Eur J Cancer 1977; 13:439 - 40; http://dx.doi.org/10.1016/0014-2964(77)90099-8; PMID: 872860
  • Juillard GJ, Boyer PJ, Niewisch H, Hom M. Distribution and consequences of cell suspensions following intralymphatic infusion. Bull Cancer 1979; 66:217 - 28; PMID: 486731
  • Juillard GJ, Boyer PJ, Snow HD. Intralymphatic infusion of autochthonous tumor cells in canine lymphoma. Int J Radiat Oncol Biol Phys 1976; 1:497 - 503; http://dx.doi.org/10.1016/0360-3016(76)90017-1; PMID: 972110
  • Juillard GJ, Boyer PJ, Yamashiro CH. A phase I study of active specific intralymphatic immunotherapy (ASILI). Cancer 1978; 41:2215 - 25; http://dx.doi.org/10.1002/1097-0142(197806)41:6<2215::AID-CNCR2820410622>3.0.CO;2-X; PMID: 77718
  • Juillard GJ, Boyer PJ, Yamashiro CH, Snow HD, Weisenburger TH, McCarthy T, et al. Regional intralymphatic infusion (ILI) of irradiated tumor cells with evidence of distant effects. Cancer 1977; 39:126 - 30; http://dx.doi.org/10.1002/1097-0142(197701)39:1<126::AID-CNCR2820390122>3.0.CO;2-5; PMID: 832227
  • Johansen P, Häffner AC, Koch F, Zepter K, Erdmann I, Maloy K, et al. Direct intralymphatic injection of peptide vaccines enhances immunogenicity. Eur J Immunol 2005; 35:568 - 74; http://dx.doi.org/10.1002/eji.200425599; PMID: 15682446
  • Heinzerling L, Basch V, Maloy K, Johansen P, Senti G, Wüthrich B, et al. Critical role for DNA vaccination frequency in induction of antigen-specific cytotoxic responses. Vaccine 2006; 24:1389 - 94; http://dx.doi.org/10.1016/j.vaccine.2005.09.018; PMID: 16225968
  • Maloy KJ, Erdmann I, Basch V, Sierro S, Kramps TA, Zinkernagel RM, et al. Intralymphatic immunization enhances DNA vaccination. Proc Natl Acad Sci USA 2001; 98:3299 - 303; http://dx.doi.org/10.1073/pnas.051630798; PMID: 11248073
  • Smith KA, Tam VL, Wong RM, Pagarigan RR, Meisenburg BL, Joea DK, et al. Enhancing DNA vaccination by sequential injection of lymph nodes with plasmid vectors and peptides. Vaccine 2009; 27:2603 - 15; http://dx.doi.org/10.1016/j.vaccine.2009.02.038; PMID: 19428867
  • Kreiter S, Diken M, Selmi A, Diekmann J, Attig S, Hüsemann Y, et al. FLT3 ligand enhances the cancer therapeutic potency of naked RNA vaccines. Cancer Res 2011; 71:6132 - 42; http://dx.doi.org/10.1158/0008-5472.CAN-11-0291; PMID: 21816907
  • Kreiter S, Selmi A, Diken M, Koslowski M, Britten CM, Huber C, et al. Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity. Cancer Res 2010; 70:9031 - 40; http://dx.doi.org/10.1158/0008-5472.CAN-10-0699; PMID: 21045153
  • Van Lint S, Goyvaerts C, Maenhout S, Goethals L, Disy A, Benteyn D, et al. Preclinical evaluation of TriMix and antigen mRNA-based antitumor therapy. Cancer Res 2012; 72:1661 - 71; http://dx.doi.org/10.1158/0008-5472.CAN-11-2957; PMID: 22337996
  • Sigel MB, Sinha YN, VanderLaan WP. Production of antibodies by inoculation into lymph nodes. Methods Enzymol 1983; 93:3 - 12; http://dx.doi.org/10.1016/S0076-6879(83)93031-8; PMID: 6346012
  • Nilsson BO, Svalander PC, Larsson A. Immunization of mice and rabbits by intrasplenic deposition of nanogram quantities of protein attached to Sepharose beads or nitrocellulose paper strips. J Immunol Methods 1987; 99:67 - 75; http://dx.doi.org/10.1016/0022-1759(87)90033-0; PMID: 3571992
  • Lehner T, Wang Y, Cranage M, Bergmeier LA, Mitchell E, Tao L, et al. Protective mucosal immunity elicited by targeted iliac lymph node immunization with a subunit SIV envelope and core vaccine in macaques. Nat Med 1996; 2:767 - 75; http://dx.doi.org/10.1038/nm0796-767; PMID: 8673922
  • Bogers WM, Bergmeier LA, Ma J, Oostermeijer H, Wang Y, Kelly CG, et al. A novel HIV-CCR5 receptor vaccine strategy in the control of mucosal SIV/HIV infection. AIDS 2004; 18:25 - 36; http://dx.doi.org/10.1097/00002030-200401020-00003; PMID: 15090826
  • Bogers WM, Bergmeier LA, Oostermeijer H, ten Haaft P, Wang Y, Kelly CG, et al. CCR5 targeted SIV vaccination strategy preventing or inhibiting SIV infection. Vaccine 2004; 22:2974 - 84; http://dx.doi.org/10.1016/j.vaccine.2004.02.050; PMID: 15356916
  • Kawabata S, Miller CJ, Lehner T, Fujihashi K, Kubota M, McGhee JR, et al. Induction of Th2 cytokine expression for p27-specific IgA B cell responses after targeted lymph node immunization with simian immunodeficiency virus antigens in rhesus macaques. J Infect Dis 1998; 177:26 - 33; http://dx.doi.org/10.1086/513811; PMID: 9419166
  • Klavinskis LS, Bergmeier LA, Gao L, Mitchell E, Ward RG, Layton G, et al. Mucosal or targeted lymph node immunization of macaques with a particulate SIVp27 protein elicits virus-specific CTL in the genito-rectal mucosa and draining lymph nodes. J Immunol 1996; 157:2521 - 7; PMID: 8805653
  • Lehner T, Bergmeier LA, Tao L, Panagiotidi C, Klavinskis LS, Hussain L, et al. Targeted lymph node immunization with simian immunodeficiency virus p27 antigen to elicit genital, rectal, and urinary immune responses in nonhuman primates. J Immunol 1994; 153:1858 - 68; PMID: 7519218
  • Lehner T, Mitchell E, Bergmeier L, Singh M, Spallek R, Cranage M, et al. The role of gammadelta T cells in generating antiviral factors and beta-chemokines in protection against mucosal simian immunodeficiency virus infection. Eur J Immunol 2000; 30:2245 - 56; http://dx.doi.org/10.1002/1521-4141(2000)30:8<2245::AID-IMMU2245>3.0.CO;2-7; PMID: 10940916
  • Barth RJ Jr., Fisher DA, Wallace PK, Channon JY, Noelle RJ, Gui J, et al. A randomized trial of ex vivo CD40L activation of a dendritic cell vaccine in colorectal cancer patients: tumor-specific immune responses are associated with improved survival. Clin Cancer Res 2010; 16:5548 - 56; http://dx.doi.org/10.1158/1078-0432.CCR-10-2138; PMID: 20884622
  • Czerniecki BJ, Koski GK, Koldovsky U, Xu S, Cohen PA, Mick R, et al. Targeting HER-2/neu in early breast cancer development using dendritic cells with staged interleukin-12 burst secretion. Cancer Res 2007; 67:1842 - 52; http://dx.doi.org/10.1158/0008-5472.CAN-06-4038; PMID: 17293384
  • Fadul CE, Fisher JL, Hampton TH, Lallana EC, Li Z, Gui J, et al. Immune response in patients with newly diagnosed glioblastoma multiforme treated with intranodal autologous tumor lysate-dendritic cell vaccination after radiation chemotherapy. J Immunother 2011; 34:382 - 9; http://dx.doi.org/10.1097/CJI.0b013e318215e300; PMID: 21499132
  • Lesimple T, Neidhard EM, Vignard V, Lefeuvre C, Adamski H, Labarrière N, et al. Immunologic and clinical effects of injecting mature peptide-loaded dendritic cells by intralymphatic and intranodal routes in metastatic melanoma patients. Clin Cancer Res 2006; 12:7380 - 8; http://dx.doi.org/10.1158/1078-0432.CCR-06-1879; PMID: 17189411
  • Lesterhuis WJ, de Vries IJ, Schreibelt G, Lambeck AJ, Aarntzen EH, Jacobs JF, et al. Route of administration modulates the induction of dendritic cell vaccine-induced antigen-specific T cells in advanced melanoma patients. Clin Cancer Res 2011; 17:5725 - 35; http://dx.doi.org/10.1158/1078-0432.CCR-11-1261; PMID: 21771874
  • Schwaab T, Schwarzer A, Wolf B, Crocenzi TS, Seigne JD, Crosby NA, et al. Clinical and immunologic effects of intranodal autologous tumor lysate-dendritic cell vaccine with Aldesleukin (Interleukin 2) and IFN-alpha2a therapy in metastatic renal cell carcinoma patients. Clin Cancer Res 2009; 15:4986 - 92; http://dx.doi.org/10.1158/1078-0432.CCR-08-3240; PMID: 19622576
  • Yi Q, Szmania S, Freeman J, Qian J, Rosen NA, Viswamitra S, et al. Optimizing dendritic cell-based immunotherapy in multiple myeloma: intranodal injections of idiotype-pulsed CD40 ligand-matured vaccines led to induction of type-1 and cytotoxic T-cell immune responses in patients. Br J Haematol 2010; 150:554 - 64; http://dx.doi.org/10.1111/j.1365-2141.2010.08286.x; PMID: 20618329
  • Weber J, Boswell W, Smith J, Hersh E, Snively J, Diaz M, et al. Phase 1 trial of intranodal injection of a Melan-A/MART-1 DNA plasmid vaccine in patients with stage IV melanoma. J Immunother 2008; 31:215 - 23; http://dx.doi.org/10.1097/CJI.0b013e3181611420; PMID: 18481391
  • Ribas A, Weber JS, Chmielowski B, Comin-Anduix B, Lu D, Douek M, et al. Intra-lymph node prime-boost vaccination against Melan A and tyrosinase for the treatment of metastatic melanoma: results of a phase 1 clinical trial. Clin Cancer Res 2011; 17:2987 - 96; http://dx.doi.org/10.1158/1078-0432.CCR-10-3272; PMID: 21385924
  • Weber JS, Vogelzang NJ, Ernstoff MS, Goodman OB, Cranmer LD, Marshall JL, et al. A phase 1 study of a vaccine targeting preferentially expressed antigen in melanoma and prostate-specific membrane antigen in patients with advanced solid tumors. J Immunother 2011; 34:556 - 67; http://dx.doi.org/10.1097/CJI.0b013e3182280db1; PMID: 21760528
  • Adamina M, Rosenthal R, Weber WP, Frey DM, Viehl CT, Bolli M, et al. Intranodal immunization with a vaccinia virus encoding multiple antigenic epitopes and costimulatory molecules in metastatic melanoma. Mol Ther 2010; 18:651 - 9; http://dx.doi.org/10.1038/mt.2009.275; PMID: 19935776
  • Spaner DE, Astsaturov I, Vogel T, Petrella T, Elias I, Burdett-Radoux S, et al. Enhanced viral and tumor immunity with intranodal injection of canary pox viruses expressing the melanoma antigen, gp100. Cancer 2006; 106:890 - 9; http://dx.doi.org/10.1002/cncr.21669; PMID: 16404742
  • Arbes SJ Jr., Gergen PJ, Elliott L, Zeldin DC. Prevalences of positive skin test responses to 10 common allergens in the US population: results from the third National Health and Nutrition Examination Survey. J Allergy Clin Immunol 2005; 116:377 - 83; http://dx.doi.org/10.1016/j.jaci.2005.05.017; PMID: 16083793
  • The International Study of Asthma and Allergies in Childhood (ISAAC) Steering Committee. Worldwide variation in prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and atopic eczema: ISAAC. Lancet 1998; 351:1225 - 32; http://dx.doi.org/10.1016/S0140-6736(97)07302-9; PMID: 9643741
  • Verlato G, Corsico A, Villani S, Cerveri I, Migliore E, Accordini S, et al. Is the prevalence of adult asthma and allergic rhinitis still increasing? Results of an Italian study. J Allergy Clin Immunol 2003; 111:1232 - 8; http://dx.doi.org/10.1067/mai.2003.1484; PMID: 12789222
  • Wüthrich B, Schindler C, Medici TC, Zellweger JP, Leuenberger P, SAPALDIA (Swiss Study on Air Pollution and Lung Diseases in Adults) Team. IgE levels, atopy markers and hay fever in relation to age, sex and smoking status in a normal adult Swiss population. Int Arch Allergy Immunol 1996; 111:396 - 402; http://dx.doi.org/10.1159/000237398; PMID: 8957114
  • Pajno GB, Barberio G, De Luca F, Morabito L, Parmiani S. Prevention of new sensitizations in asthmatic children monosensitized to house dust mite by specific immunotherapy. A six-year follow-up study. Clin Exp Allergy 2001; 31:1392 - 7; http://dx.doi.org/10.1046/j.1365-2222.2001.01161.x; PMID: 11591189
  • Möller C, Dreborg S, Ferdousi HA, Halken S, Høst A, Jacobsen L, et al. Pollen immunotherapy reduces the development of asthma in children with seasonal rhinoconjunctivitis (the PAT-study). J Allergy Clin Immunol 2002; 109:251 - 6; http://dx.doi.org/10.1067/mai.2002.121317; PMID: 11842293
  • Johansen P, Senti G, Martinez Gomez JM, Storni T, von Beust BR, Wüthrich B, et al. Toll-like receptor ligands as adjuvants in allergen-specific immunotherapy. Clin Exp Allergy 2005; 35:1591 - 8; http://dx.doi.org/10.1111/j.1365-2222.2005.02384.x; PMID: 16393325
  • Johansen P, Senti G, Martínez Gómez JM, Wüthrich B, Bot A, Kündig TM. Heat denaturation, a simple method to improve the immunotherapeutic potential of allergens. Eur J Immunol 2005; 35:3591 - 8; http://dx.doi.org/10.1002/eji.200535076; PMID: 16285011
  • Martínez-Gómez JM, Johansen P, Erdmann I, Senti G, Crameri R, Kündig TM. Intralymphatic injections as a new administration route for allergen-specific immunotherapy. Int Arch Allergy Immunol 2009; 150:59 - 65; http://dx.doi.org/10.1159/000210381; PMID: 19339803
  • Martínez-Gómez JM, Johansen P, Rose H, Steiner M, Senti G, Rhyner C, et al. Targeting the MHC class II pathway of antigen presentation enhances immunogenicity and safety of allergen immunotherapy. Allergy 2009; 64:172 - 8; http://dx.doi.org/10.1111/j.1398-9995.2008.01812.x; PMID: 19076537
  • Mohanan D, Slütter B, Henriksen-Lacey M, Jiskoot W, Bouwstra JA, Perrie Y, et al. Administration routes affect the quality of immune responses: A cross-sectional evaluation of particulate antigen-delivery systems. J Control Release 2010; 147:342 - 9; http://dx.doi.org/10.1016/j.jconrel.2010.08.012; PMID: 20727926
  • Senti G, Prinz Vavricka BM, Erdmann I, Diaz MI, Markus R, McCormack SJ, et al. Intralymphatic allergen administration renders specific immunotherapy faster and safer: a randomized controlled trial. Proc Natl Acad Sci USA 2008; 105:17908 - 12; http://dx.doi.org/10.1073/pnas.0803725105; PMID: 19001265
  • Malling H, Blom L, Poulsen B, Poulsen L, Witten M. . Is intralymphatic specific immunotherapy with grass pollen allergen ready for clinical use?European Academy of Allergy and Clinical Immunology Congress. . Geneva: EAACI, 2012:S1595.
  • Siegrist CA. The Immunology of Vaccination. In: Plotkin SA, Orenstein WA, Offit PA, eds. Vaccines. Philadelphia: Elsevier Inc., 2008:17-36.
  • Guery JC, Galbiati F, Smiroldo S, Adorini L. Selective development of T helper (Th)2 cells induced by continuous administration of low dose soluble proteins to normal and beta(2)-microglobulin-deficient BALB/c mice. J Exp Med 1996; 183:485 - 97; http://dx.doi.org/10.1084/jem.183.2.485; PMID: 8627161
  • Senti G, Crameri R, Kuster D, Johansen P, Martinez-Gomez JM, Graf N, et al. Intralymphatic immunotherapy for cat allergy induces tolerance after only 3 injections. J Allergy Clin Immunol 2012; 129:1290 - 6; http://dx.doi.org/10.1016/j.jaci.2012.02.026; PMID: 22464647

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.