12,020
Views
42
CrossRef citations to date
0
Altmetric
Reviews

Noninvasive vaccination against infectious diseases

, , &
Pages 1717-1733 | Received 01 Sep 2017, Accepted 30 Mar 2018, Published online: 17 May 2018

ABSTRACT

The development of a successful vaccine, which should elicit a combination of humoral and cellular responses to control or prevent infections, is the first step in protecting against infectious diseases. A vaccine may protect against bacterial, fungal, parasitic, or viral infections in animal models, but to be effective in humans there are some issues that should be considered, such as the adjuvant, the route of vaccination, and the antigen-carrier system. While almost all licensed vaccines are injected such that inoculation is by far the most commonly used method, injection has several potential disadvantages, including pain, cross contamination, needlestick injury, under- or overdosing, and increased cost. It is also problematic for patients from rural areas of developing countries, who must travel to a hospital for vaccine administration. Noninvasive immunizations, including oral, intranasal, and transcutaneous administration of vaccines, can reduce or eliminate pain, reduce the cost of vaccinations, and increase their safety. Several preclinical and clinical studies as well as experience with licensed vaccines have demonstrated that noninvasive vaccine immunization activates cellular and humoral immunity, which protect against pathogen infections. Here we review the development of noninvasive immunization with vaccines based on live attenuated virus, recombinant adenovirus, inactivated virus, viral subunits, virus-like particles, DNA, RNA, and antigen expression in rice in preclinical and clinical studies. We predict that noninvasive vaccine administration will be more widely applied in the clinic in the near future.

This article is part of the following collections:
Key Issues in Contemporary Vaccinology and Immunotherapy

Introduction

Although some infectious diseases, including malaria, meningitis, diphtheria, hepatitis B, tetanus, polio, measles, mumps, and rubella, are now controlled in developed countries due to the development of vaccines and good management of healthcare, there is still a need to improve all healthcare systems and reduce the economic disparities between developed and developing countries.Citation1 Several outbreaks in developing countries of infectious diseases, such as HIV/AIDS, malaria, tuberculosis, dengue, influenza, and other respiratory diseases, occur each year and are responsible for more than 54% of total mortality. In developing countries, strategies are urgently needed to prevent these diseases as are new approaches to developing specific drugs and protective vaccines, which are currently unavailable for some diseases. The available vaccines, such as for hepatitis B and measles, elicit specific humoral and cellular immunity and protect both immunized individuals and their neighbors.Citation2-4 However, these diseases are still seriously threatening to human life, especially in developing countries.Citation5 WHO vaccination campaigns have decreased mortality, but improvements in vaccine administration and storage are still needed.

The overwhelming number of vaccinations administered worldwide over the last half century have been by injection. Such vaccinations have been responsible for preventing millions of deaths per year from infectious diseases. Nevertheless, there are reasons to consider routes of vaccination not involving injection. Of the possible vaccination methods, injection has the advantages of low cost and quick delivery, and has an excellent long-term record for safety in use across billions of doses administered worldwide over more than the past half-century.Citation6 However, most needle-based vaccines require a cold chain, which is expensive, due to the need for cold storage rooms and cold transportation to maintain the vaccine at the proper temperature.Citation7 Temperature changes could destroy freezing- or heat-sensitive vaccine vials, which would then need to be destroyed. A study in Bangladesh demonstrated that, while a cold chain could be maintained during storage of polio vaccine at the district level and at health facilities, there were temperature fluctuations during transport. Temperatures >8°C (with a maximum of 21°C) were detected in 5 of the 23 refrigerators and 3 of 6 transported cold boxes when the vaccines were carried from the district to local depots. Moreover, 17% of the vaccine carriers could not reach some remote destinations. In addition, in African, Latin American, and some Asian countries, the electrical supply is not reliable and will require extra investment in generators or conversion to alternative sources, such as solar energy, to provide consistent cold chain storage.Citation8

Furthermore, hypodermic injection is not easy to perform at home by the patients themselves and requires the help of trained nurses.Citation9 Moreover, hypodermic injection may result in pain for patients, thus potentially leading to needle phobia, especially in children.Citation10,Citation11 The improper disposal and reuse of syringes contaminated with blood-borne pathogens are also essential safety concerns, especially in developing countries.Citation12,Citation13

In recent decades, needle-free vaccine delivery systems have been developed to improve safety, especially to healthcare providers and the community, after concerns regarding pandemics and bioterrorism. Oral, intranasal (i.n.), and transcutaneous vaccinations are promising noninvasive immunization methods that stimulate mucosal immunity, which is one of the most important components of the immune response. Mucosal immunity not only protects against respiratory infections, such as influenza, but also against systemic infections, such as tuberculosis (TB) and HIV.Citation14 Vaccine delivered directly to the mucosal surface is rapidly and widely distributed and enhances mucosal, cellular, and humoral immune responses.Citation15 I.n. vaccination against respiratory and gastrointestinal pathogens offers several theoretical and practical benefits, such as avoiding the need for extensive purification from bacteria and significantly reducing the cost of training healthcare workers. Furthermore, noninvasive vaccination eliminates needlestick injuries and exposure of health workers to blood-borne pathogens, such as HIV and hepatitis B, and there is no need to dispose of large quantities of needles, which is a difficult problem in developing countries.Citation16 In contrast to mucosal immunization, transcutaneous vaccines are not degraded by enzymes or digestive acid,Citation17 and microneedle patches are easy to carry during travel. In fact, beyond the application in vaccine delivery, microneedle-mediated vaccination also induces more efficient immunity, and it has been widely used in the treatment of diabetes, Alzheimer's diseases, seborrheic keratosis, and tumors.Citation18-21

Here we describe advances in oral, i.n., and transcutaneous vaccination at different stages of development, including preclinical and clinical trials and licensed vaccines. We focus on vaccine technologies that are based on noninvasive immunization with live attenuated virus, recombinant virus, inactivated virus, viral subunits, virus-like particles, DNA, RNA, and antigen expression in rice.

Noninvasive vaccines induce mucosal and systemic immunity

The epithelial coating of mucous membranes constitutes the largest immunologic organ, which has a greater probability of infection if the microorganism crosses at least one of the body's protective mucosal barriers. These barriers are the respiratory tract, gut, genital tract, conjunctiva, and urinary system. The protection of mucosal membranes is conferred by the humoral and T cell responses as well as the innate immune response, and this system is collectively known as mucosa-associated lymphoid tissue (MALT).Citation22 MALT immunization is similar to systemic immunization, which depends on efficient antigen sampling and uptake, antigen presentation by antigen-presenting cells (APCs), and the production of effector and memory B and T cells.Citation23 MALT can be subdivided into gut-associated lymphoid tissue (GALT), which includes lymph nodes, Peyer's patches, and isolated lymphoid follicles, and nasopharyngeal-associated lymphoid tissue (NALT), which includes tonsils/adenoids, inducible bronchus-associated lymphoid tissue, and lymph nodes.

Unlike systemic immunization, mucosal immunization depends on the secretion of IgA by plasma cells.Citation24 Locally, secreted IgA (sIgA) binds to the Ig receptor (pIgR) at the basolateral surface of mucosal epithelial cells and is then actively transported into the lumen by transcytosis,Citation25,Citation26 where it regulates viral and bacterial infections at the oral and i.n. mucosal surface. The sIgA antibodies bind to pathogens to inhibit their interaction with, and uptake by, epithelial cells but deliver the pathogens to antigen-presenting cells (APCs).Citation27 In GALT locations, the precursors of IgA plasma cells are generated in localized zones of organized lymphoid induction sites and gut lymph nodes.Citation28,Citation29 These structures are found in the small intestine and appendix, where the B cells are activated at the germinal centers (GCs). The generation and maturation of IgA-producing B cells are promoted by follicle DCs, T follicular helper cells, and macrophages, which secrete cytokines, such as TGF-β, IL-10, and IL-4,Citation30 and help the B cells to generate long-lasting memory cells with high levels of somatic mutation.Citation31,Citation32 The NALT in humans is localized in the salivary glands, the paired palatine and tubal tonsils, and the unpaired pharyngeal and lingual tonsils and has similar functions as GALT. The tonsils consist of follicular germinal centers, the extrafollicular areas, the mantle zones, and the reticular crypt epithelium on the surface. The epithelium contains M cells, DCs, naïve B and T cells, and memory B cells. Unlike GALT, the tonsils generate plasma cells with a production of 70% IgG and 30% IgA. However, the paired tubal tonsils and unpaired pharyngeal tonsils produce sIgA, which blocks entry into the epithelia cells. Like GALT, the germinal centers are associated with somatic mutation and differentiation into B memory cells.Citation33

In addition, it has been observed that mucosal and transcutaneous immunization induce systemic immune responses that are further enhanced by activated dendritic cell (DC) migration from the mucosa to the lymph nodes and spleen.Citation27 It has also been observed that, with the help of L-selectin on the cell surface,Citation34 systemic IgG B cells migrate into the mucosa from bone marrow, and, vice versa, that induced antigen-specific T and B cells leave the Peyer's patches via the efferent lymphatics through thoracic duct lymph and are disseminated to the systemic circulation. They then enter the mucosa of the gut, respiratory system, salivary and mammary glands, and reproductive tract.Citation35,Citation36

Oral vaccination preclinical applications

Oral vaccination is the best method for vaccine administration because of the ease of regulatory compliance and low cost. Oral immunization is a standard needle-free technique that has been used especially in vaccine campaigns against influenza (children), poliomyelitis, rotavirus, typhoid fever, cholera, and other diseases.Citation22 However, only a few vaccines have an oral formulation, since there are issues that may affect protective immune responses. For example, antigen should travel intact to the lumen of the intestine without degradation by the acidic environment of the stomach.Citation37,Citation38 Otherwise, the antigen concentration could diminish from levels capable of inducing an immune response to an anergy response. However, a good vaccine like the poliomyelitis vaccine passes through the digestive tract to the intestine, where the M cells in the Peyer's patches (PPs) of the GALTCitation37 lack a mucus layer on their apical side and efficiently transport oral vaccines to APCs by transcytosis,Citation27,Citation39 In fact, APCs are essential cells for the initiation of local and systemic immunity; therefore, developing vaccine carriers targeting APCs is a good strategy to improve the immune response.Citation3

Oral delivery has been tested in development of viral and bacterial vaccines that were attenuated by repeated passages in tissue culture or by chemical mutagenesis. This approach was first demonstrated in a mouse model by sublingual immunization with a live attenuated Japanese encephalitis virus vaccine and a recombinant modified vaccinia virus, which induced virus-specific IFN-γ+ CD4+ and CD8+ T cells. However, IL-17+ CD4+ T cells were also increased after sublingual immunization.Citation40

Intriguingly, DNA vaccines delivered by live attenuated Salmonella typhimurium (JOL911) effectively protected mice from lethal infection by H1N1 influenza virus, while i.n. administration did not. However, virus copy numbers in the lungs were lower following oral and i.n. immunization compared with the lungs of the PBS-vaccinated control group.Citation41 In another study, attenuated S. typhimurium was used to orally deliver a Trichinella spiralis paramyosin DNA vaccine to mice. The orally vaccinated mice were effectively protected from T. spiralis infection, and their antibody responses showed significant mucosal sIgA and systemic IgG2a and a significant increase in Th1 (IFN-γ, IL-2) and Th2 (IL-4, -5, -6, and -10) cytokines.Citation42 Oral immunization with antigens carried by engineered viruses has demonstrated that GALT elicits both humoral and cellular immune responses that protect against several infectious diseases. The most frequently used engineered virus is adenovirus, which has been used in phase I clinical trials. Several reports have demonstrated that adenovirus vaccine can be administered orally to prevent different kinds of infection. Lubeck developed human adenovirus type 7 (Ad7) and type 4 (Ad4) vaccines containing hepatitis B surface antigen. After oral immunization of chimpanzees with these two vaccines, the animals generated significant antibody responses and effective protection against hepatitis B virus.Citation43 In another study, skunks and foxes were vaccinated with an Ad5 vaccine expressing the rabies glycoprotein gene. The vaccine was instilled into the mouth cavity, resulting in a 100% survival rate for street rabies virus-challenged animals.Citation44 Furthermore, studies with mice orally vaccinated with a replication-deficient adenovirus (Rad68) vaccine containing the measles virus nucleocapsid protein generated a significant splenic cytotoxic T cell response (in 70% of the mice) and antibody response (in 89% of the mice).Citation45 Oral administration of adenovirus vaccine also protected the mice from HIV, Ebola, influenza virus, and botulism.Citation46-49 In another study by Lin et al., vaccination with live attenuated gastroenteritis virus incorporating CpG DNA in pigs enhanced the IgA level in the intestinal tract and the IgG level in the serum after oral immunization.Citation50

Currently, influenza vaccines are administered intramuscularly or intradermally, which induces mainly humoral responses against hemagglutinin (HA) and neuraminidase (NA) proteins. These proteins are polymorphic, especially in the exposed domains subject to the immune system.Citation51,Citation52 Nevertheless, these routes of immunization are less effective in stimulating mucosal immunity,Citation53,Citation54 and a split-flu vaccine in combination with the adjuvant methylglycol chitosan and/or CRX601 administered sublingually was recently found to improve the systemic and mucosal immune responses equivalently or to a greater extent than intramuscular vaccination.Citation55

Subunit vaccines based on virus-like particles (VLPs) that self-assemble from viral structural proteins and related antigens is another technology used to stimulate GALT and induce protection.Citation56 This type of vaccine demonstrated that oral immunization with recombinant Baccillus subtilis expressing cholera toxin B subunits and Helicobacter pylori urease B spores contributes to a reduction in the H. pylori load.Citation57

As mentioned above, one of the problems of vaccination in developing countries is maintaining a cold chain to preserve vaccines. To solve this problem, Borde et al. developed a liquid-killed, multivalent whole-cell-plus-enterotoxin-B-subunit oral vaccine against enterotoxigenic E. coli that induced an effective immune response in mice.Citation58 This vaccine is given together with dmLT (an enterotoxin-derived adjuvant) as a dry-powder vaccine formulation that is especially suitable for low-income countries.

Another problem for vaccination is the acidic conditions of the gastrointestinal tract, which could degrade protein, DNA, and polysaccharide vaccines, leading to weak immune responses and poor protection against pathogens. New carriers are needed to design available vaccine payloads, with effective release within their residence time in the small intestine and optimization of the adjuvant capabilities of the delivery vehicles,Citation41 and some researchers have studied this kind of delivery vehicle. For example, Yagnik, et al. constructed a recombinant Lactococcus lactis bacterium that expressed protein A of Shigella dysenteriae type 1 in its outer membrane, which elicited a higher immune response after oral administration than by the i.n. route.Citation59 In another study, Oliveira et al. delivered a plasmid DNA vaccine expressing the Rho1 GTP protein of Schistosoma mansoni with chitosan nanoparticles. In vivo experiments suggested that oral administration increases the expression of modulatory IL-10 and thereby reduces liver pathology.Citation60 Moreover, green-synthesis silver nanoparticles formulated with an H5N1 DNA vaccine generated effective antibody and cellular immunity in chicken against influenza.Citation61 In fact, oral vaccine administration has several protective effects besides the control of infectious diseases. For example, the cholera toxin B subunit, a component of a licensed oral cholera vaccine, increased mucosal healing in the colon after oral administration.Citation62 Also, hepcortespenlisimut-L, a drug used as a liver cancer vaccine, has proven to be safe, effective, and fast-acting against hepatocellular carcinoma following oral administration.Citation63

Interestingly, a transgenic rice-based vaccine has been developed, which has proven to be safe and resistant to the acidic environment in the stomach.Citation64 It has been demonstrated that rice-based allergic vaccines as well as cholera and diarrhea vaccines can be delivered efficiently by oral administration.Citation65-67Taken together, these results suggest that oral vaccine administration has great potential for protection against different diseases.

Clinical trials for oral vaccines

Several clinical trials for oral vaccines have been conducted and are listed in . However, these vaccines have not been approved by the US FDA. Rotarix and RotaTeq are two vaccines that have been internationally approved by WHO; however, they have been shown to be less effective (in 40–70% of subjects) in third world countries.Citation68 The live attenuated rotavirus vaccine Rotavin-M1 was evaluated for safety and immunogenicity, and the IgA seroconversion rate of Rotavin-M1 was found to be comparable to the licensed vaccine Rotarix.Citation69 However, infants who received Rotavin-M1 shed virus in their stool at a lower frequency (44–48%) than those who received Rotarix (65%). More infants are expected to be enrolled in subsequent trials.

Table 1. Clinical trials for oral immunization.

Another vaccine in clinical trials is designed against enterotoxigenic E. coli (ETEC), which is endemic in developing countries and may produce an infectious diarrhea. Chen et al. conducted a phase 1 clinical trial to demonstrate the safety and immunogenicity of the peru-15pCTB vaccine against ETEC infection. This study showed that antitoxin antibody seroconversion increased fourfold compared with baseline. Furthermore, the vaccine was safe up to 1 × 1010 CFU.Citation70 Recently, an oral recombinant adenovirus influenza vaccine induced significant increases in haemagglutination inhibition and high titers of microneutralization antibodies with only mild adverse effects.Citation71 Moreover, Euvichol is an inactivated bivalent oral cholera vaccine tested during a phase I clinical trial in healthy South Korean adult males that induced a seroconversion rate of 95% and 45% for Vibrio cholera O1 and O139, respectively.Citation72

H. pylori is associated with gastritis, peptic ulcer, and gastric adenocarcinoma and is present in the gastric mucosa in at least half of the world population. Until now, there has been no effective vaccine. However, an oral recombinant H. pylori vaccine substantially reduced infection by this bacterium in clinical trials that had enrolled 4464 children from China.Citation73

One more vaccine assessed in a phase I clinical trial is an adenovirus vaccine that expresses both an avian influenza A hemagglutinin and a TLR3 ligand. This vaccine exhibited cytotoxic T cell responses and IFN-γ production that were similar to the placebo group.Citation74 Efficient immunogenicity and fewer adverse effects were also found in these clinical trials. However, more participants are needed to further evaluate these vaccines before they receive final approval.

Licensed oral vaccines

The poliomyelitis is a disease caused by three poliovirus serotypes (P1, P2, and P3). The virus enters through the mouth, goes to the gastrointestinal tract, then to the lymph nodes and may travel to the central nervous system, where the virus replicates in the motor neural cells, resulting in motor neuron destruction causing muscle weakness and paralysis.Citation75 The first immunotherapy was developed in the 1950s by Hammon et al, who used immunoglobulins of individuals recovered from poliovirus infection and injected into patients with active disease. This passive immunization was 80% effective reducing the effects of poliomyelitis, but there were not enough plasma to stop the pandemic.Citation76

The first candidate polio vaccine was developed by Hilary Koprowski and colleagues. This poliovirus vaccine was attenuated in cotton rats with low pathogenicity in rhesus monkeys. It was first tested in 20 healthy individuals, and they were all seroconverted and did not get sick.Citation77 The first licensed vaccine (inactivated poliovirus vaccine, IPV) contains P1, P2 and P3 serotypes, which were grown in Vero cells (monkey kidney cells) and inactivated with formaldehyde. The volunteers who were immunized showed seroconversion with 3 doses.Citation78 This vaccine was developed by Sack et al., and it was licensed for serotypes P1 and P2 in 1961 and type 3 in 1963.Citation79 The oral poliovirus vaccine (OPV) was the second vaccine, composed of the subtypes P1, P2 and P3 after the repeated passage of the virus in tissue cultures of cynomolgus monkey kidney at subphysiological temperatures. The subtype viruses are able to infect cells in the gut but is unable to replicate in the neuron system. The vaccine was developed by Sabin et al., and it was licensed in 1962. OPV vaccine was tested in several countries (Netherlands, Mexico, Japan, USSR, Brazil, Cuba, etc.) before its license.Citation80 Due to its low cost, it became the most used polio vaccine worldwide. However, OPV can cause vaccine-associated paralytic poliomyelitis (VAPP) especially in developing countries with a risk of 4.7 cases per million births. VAPP were associated with subtype 2 poliovirus.Citation81 This vaccine was discontinued in the USA in 2000. The VAPP has not been observed using the IPV. OPV is used in many countries. However, WHO recommends that all countries using only OPV add at least 1 dose of IPV to the schedule.Citation82 The IPV and OPV are licensed in the USA but only IPV is currently used. The most recent version of IPV contains three serotypes of polio vaccine viruses, but grown single component in Vero cells, inactivated with formaldehyde, and has trace amounts of antibiotics as a preservative. Both polio vaccines have saved millions of lives and have prevented deformities, which result in significant high cost in health care especially in developing countries.

Adenovirus types 4 and 7 cause acute respiratory disease, which is a threat to the health of US military personnel.Citation83 A vaccine for adenovirus types 4 and 7 (BL 125296/0) is orally administered and after 26 days in a clinical trial showed a seroconversion rate of 94.5% and 93.8% against types 4 and 7, respectively.Citation84 The protective effect of this vaccine was associated with serotype-specific neutralizing antibodies. While there were adverse effects, such as nasal congestion, cough, sore throat, headache, abdominal pain, arthralgia, nausea, and diarrhea, there was not a significant difference from the placebo group. Vaxchora is an FDA-approved oral vaccine for individuals of ages 18–64 years traveling to cholera-endemic areas. This vaccine was 90.3% effective after 10 days and 79.5% effective after 90 days against V. cholerae O1, and the immune mechanism is associated with vibriocidal antibody titers.Citation85

Rotarix™, an orally administered vaccine, is used widely to protect from gastroenteritis and reduce its occurrence after hospitalization.Citation86,Citation87 A clinical study showed that after one month of a two-dose series, 86.5% of 787 recipients of Rotarix were seroconverted compared with 6.7% of placebo recipients. A pentavalent vaccine, RotaTeq, was also shown to protect against rotavirus gastroenteritis at different levels of severity by increased IgA seroconversion.Citation88,Citation89 Both Rotarix and RotaTeq are more suitable for protecting against severe gastroenteritis (effective for 90% of patients) while less suitable against mild infection (effective for 60–75% of patients).Citation90 However, it has been documented that the use of RotaTeq and Rotarix has been associated with the development at a very low rate of intussusception (∼1:40,000 doses), and shedding from the vaccinee's stools, which may result in the infection of immunocompromised individuals.Citation27

Salmonella typhi causes typhoid fever, an acute, febrile, enteric disease, which continues to be important in many parts of the world.Citation91 Vivotif is another vaccine against gut pathogens and is administered by the oral route and is indicated for immunization against S. typhi for adults and children older than 6 years. This capsule vaccine should be administered in four doses to induce a broad immune response, mainly including serum IgG and IgA responses, mucosal antibodies, CD4+ T cells, and CD8+ cytotoxic T cells.Citation22,Citation92-94 However, most of these licensed vaccines are not recommended for infants due to the lack of safety data for attenuated virus vaccines, and only rotavirus vaccines are currently used in infants.Citation27

The oral killed-cholera vaccines, Shanchol and Dukoral, are safe, immunogenic, effective in the clinic, and are recommended by WHO. However, they are not available in the US as of this writing. It was found that vibriocidal antibodies against V. cholera O1 Inaba, V. cholerae O1 Ogawa, and V. cholerae O139 were effectively induced in adults, toddlers, and younger children.Citation95 In addition, Dukoral is safe and immunogenic in Peruvian and North American volunteers.Citation96,Citation97 Sriskyi et al. showed that TLR–MyD88 signaling may mediate immune responses to the Dukoral vaccine.Citation98 Furthermore, antigen-specific memory B-cell responses were not detected in Dukoral-treated individuals, which explains the brief period of protection conferred by this vaccine.Citation99

Preclinical evaluation of intranasal vaccination

Several diseases, such as influenza and pneumococcal disease, threaten human life and are transmitted by the nasal route. Nasal tissue is also an excellent route for vaccine administration, which has the advantage of requiring lower doses and without exposure to extreme pH or digestive enzymes. Separate from the main salivary glands and tonsil tissue, there are locally draining lymph nodes that lie under the respiratory epithelium of the nasal cavity, where antigens are also transported to DCs, macrophages, and B cells by M cells. B lymphoblasts migrate to and proliferate in the germinal centers where mucosal sIgA and systemic IgG are induced.Citation100 In parallel, mature DCs bearing antigens migrate to the follicular B cell zone and interfollicular T cell zone and activate T cells and B lymphoblasts by presenting antigens to them. The activated T and B cells then enter the bloodstream to initiate systemic immunity at distant sites.Citation101 Therefore, i.n. immunization can induce potent immunity, producing effective protection against infectious disease while helping to achieve widespread vaccination in developing countries.

Studies of i.n. administration of vaccines have been conducted using live attenuated influenza vaccines, which have generated protective immune responses. Fan et al., demonstrated in rhesus macaques that i.n. vaccination with cold-attenuated H5N1 virus induces neutralizing antibodies and an HA-specific CD4+ T cell immune response that is fully protective. Yang et al. also found that ferrets and mice immunized by i.n. administration with live attenuated BJ/AA ca virus produced high levels of sIgA that was protective against challenge with A/Beijing/501/2009 or A/California/07/2009 viral strains.Citation102-104 Furthermore, an engineered PR8 influenza virus was generated that expressed the receptor-binding subdomain of botulinum neurotoxin, resulting in a vaccine that protected mice from botulinum and influenza at the same time using i.n. administration.Citation105 Moreover, an attenuated live viral vaccine expressing a functional species-specific artificial microRNA (PR8-amiR-93NP) was safe and led to cross protection for mice from heterologous influenza virus strains after i.n. immunization.Citation106

Although vaccines induce protective immune responses against infectious diseases, the adjuvant helps to accelerate and enhance antigen-specific immune responses. Huang et al., combined several adjuvants, including PELC, which is a squalene-based water-in-oil-in-water emulsion stabilized by Span®85 (sorbitan trioleate), and poly(ethylene glycol)-block-poly(lactide-co-ϵ-caprolactone) (PEG-b-PLACL) together with LD-indolicidin or alum and inactivated influenza virus to formulate a vaccine that increased influenza-specific serological protection after i.n. vaccination.Citation107

In addition, i.n. vaccines against other viruses and bacteria have been developed. For example, when used for i.n. immunization, an rAd5-based vaccine combined with 2.0 × 108 IFU Ad5-IFNα as adjuvant facilitated the development of robust specific antibodies and increased survival in guinea pigs exposed to experimental Ebola virus.Citation108 Malley et al. showed that delivery of an inactivated non-encapsulated pneumococcal vaccine and cholera toxin as adjuvant elicited protection against nasopharyngeal colonization and invasive disease from encapsulated pneumococci.Citation109 Moreover, the combined immunization of formalin-inactivated influenza vaccine and cholera toxin significantly reduced the death rate in mice after lethal infection by influenza A virus followed by group A streptococci. These results showed the importance of preventing secondary bacterial infections after prior non-lethal influenza exposure.Citation110

It was also demonstrated that both i.n. and sublingual administration of inactivated polio vaccines contributes to the generation of polio-specific serum IgA in saliva, fecal extracts, and intestinal tissues and to IgA-producing B cells in mouse spleens. The level of polio-specific IgG induced by mucosal immunity was much higher than that induced by the intramuscular route.Citation111 Interestingly, Harakuni et al. showed that the immune effects of i.n. administration on the mucosa against an unrelated Japanese encephalitis virus are better than those resulting from administration with oral and transcutaneous immunization.Citation112 These results suggest that it is necessary to develop a more suitable method of noninvasive administration.

Likewise, subunit vaccines have been developed for delivery by i.n. administration, which was found to protect mice from the threat of Yersinia pestis. For example, Arnaboldi et al. developed a tobacco mosaic virus (TMV)-based delivery platform in which i.n. immunization of a subunit vaccine consisting of Y. pestis virulence factors, including F1 and LcrV, with TMV significantly reduced the morbidity and mortality of Y. pestis, while subunit vaccines lacking TMV did not.Citation113 Other i.n. vaccine candidates have been tested with encouraging results. One study showed that a lipopeptide complex including a conserved extracellular domain of matrix protein 2 (M2e) of influenza A virus induced the generation of IgA and IgG2b antibodies and reduced the mortality of influenza-challenged mice without adjuvant.Citation114 Even when tested without adjuvant, an i.n. vaccine delivery consisting of a nanometer-sized hydrogel (composed of a cationic cholesteryl group-bearing pullulan [cCHP]) with a nontoxic subunit fragment of Clostridium botulinum type A neurotoxin BoHc/A strongly increased serum IgG and sIgA antibody responses.Citation115 Furthermore, the expressed fusion protein, which included M2e and cholera toxin subunit B in a vaccine delivered by the i.n. route, induced specific IgA and IgG antibodies as well as T cell and memory B cell responses and protected mice against heterologous influenza virus.Citation116 Recombinant vaccines administered with i.n. immunization have also been widely studied. Leishmanial recombinant protein combined with cholera toxin protects mice from Leishmania infection by inducing IFN-γ production following i.n. administration.Citation117 Another study found that immunization with recombinant DnaJ (Hsp40) protein induced effective immune antibodies and the release of IL-10, IFN-γ, and IL-17A against Streptococcus pneumoniae.Citation118 Interestingly, non-pathogenic Lactococcus lactis NZ9000, expressing pneumococcal immunogenic proteins, protected against S. pneumoniae and elicited innate and adaptive immune responses.Citation119

Replication-deficient recombinant human adenovirus serotype rAd5 vectors were also constructed to express the N gene of the pneumonia virus of mice (PVM) pathogenic strain J3666. Its i.n. administration causes a PVM-specific CD8 response rather than increased serum IgG, which may result in the effective protection of mice after PVM challenge.Citation120

The effects of VLP vaccines administered by i.n. immunization have also been widely investigated. A modular murine polyomavirus-like particle delivered by i.n. immunization was used to display the group A streptococcus (GAS) antigen J8i, which significantly induced J8i-specific IgG and IgA antibodies and reduced GAS colonized in the throat.Citation121 Cai et al. showed that a combination of respiratory syncytial virus (RSV) fusion protein VLPs and glycoprotein VLPs increases protection against live RSV, with a reduction in lung viral replication and histopathology damage.Citation122 However, some adjuvants, such as Toll-like receptor 3, ricin toxin B, TLR7 and nine of its agonists, and murabutide, need to be applied to increase the immune response of VLP vaccines.Citation123-126 Following i.n. delivery of TLR7 and its nine agonists with Norwalk virus, VLPs produce a more robust and broad spectrum of immune responses than by oral administration.Citation125

Other vaccines, such as those involving DNA immunized by the i.n. route, need adjuvants, such as enterohemorrhagic E. coli-secreted proteins and IL-6, to increase the immune response.Citation127,Citation128 Currently, DNA vaccines can be delivered using several biomaterials, such as the copolymer polyethyleneimine and hollow Ag@SiO2 nanoparticles.Citation129-131 In the same way, mRNA can be transfected into both dividing and non-dividing cells without entering the nucleus, resulting in higher gene expression. Furthermore, mRNA vaccines may overcome specific mutations that frequently occur in patients. Li et al. developed a system using a cationic cyclodextrin-modified polyethylenimine 2 k conjugate with encoded HIV gp120 mRNA. This system delivered by i.n. immunization provides strong systemic and mucosal immune responses, cytokine production, and a balanced Th1/Th2/Th17 T helper cell distribution. These vaccines also overcame the nasal epithelial barrier to increase mRNA paracellular delivery.Citation132

Clinical trials for intranasal vaccines

MEDI-534 is a vaccine designed for delivery by the i.n. route that is currently undergoing a clinical trial. It combines a live attenuated respiratory syncytial virus (RSV) with parainfluenza-3 virus (PIV3), which causes lower respiratory tract illness in young children. This clinical trial demonstrated that MEDI-534 has a potential immunogenic effect in both seropositive and seronegative children.Citation133-135 Furthermore, after three doses of MEDI-534, the children were found to be 100% seroresponsive to PIV3 but only 50% to RSV. It was demonstrated that a mutation generated in the transcription termination poly A gene sequence upstream of the bPIV3N gene and the RSV fusion protein F open reading frame reduced RSV F expression. Although there was an association between this mutation and a lower rate of RSV seroconversion, a larger sample size is needed for definitive confirmation.Citation136

A live attenuated i.n.-administered pertussis vaccine, BPZE1, induced significant memory B cell responses against pertussis toxin, filamentous haemagglutinin, and pertactin in 7 colonized subjects out of 36 subjects who had received BPZE1.Citation137,Citation138 Moreover, BPZE1 had a protective effect against RSV, in part, by markedly increasing IL-17 cytokines produced by CD4+ T cells.Citation139 The peptide vaccine Vacc-4x was used for HIV therapy and generated T cell responses and mucosal and systemic humoral responses. However, different doses led to different immune responses; hence, the clinical dosage and mechanisms of immune regulation need to be further elucidated.Citation140

Atmar et al. conducted a clinical trial to assess the effect of a norovirus-like particle vaccine against norovirus, which causes epidemics and sporadic acute gastroenteritis. This study showed reduced frequencies of gastroenteritis and infection by Norwalk virus and increased IgA seroconversion.Citation141 A Norwalk VLP vaccine also enhanced the levels of specific IgG antibodies in subjects that had received a 100-μg vaccine dose and enhanced the levels of IgA antibody-secreting cells in subjects that received a 50-μg or 100-μg vaccine dose.Citation142 Despite limitations with regard to subject age, pregnancy, and health issues of the currently licensed vaccines, an i.n. proteasome-adjuvanted trivalent vaccine increased seroconversion and protection against infection.Citation143 The clinical trials that have been conducted for i.n. vaccines are listed in .

Table 2. Clinical trials for intranasal immunization.

Licensed i.n. vaccines

The US FDA-approved, i.n.-delivered, licensed vaccines are the influenza A (H1N1) 2009 monovalent pandemic flu vaccine and the FluMist® trivalent and quadrivalent seasonal flu vaccines. These vaccines activate the immune responses of individuals of ages 2–49 years, who are protected against influenza disease caused by the pandemic (H1N1) 2009 virus, while the FluMist trivalent vaccine was designed against two influenza A and one influenza B strains.Citation144 The first approved FluMist quadrivalent vaccine was intranasally delivered against H1N1 and H3N2 2009 virus and B strains by Yamagata and Victoria,Citation145,Citation146 who demonstrated that the quadrivalent and trivalent vaccines are safe and immunogenic.Citation146 Vaccination with FluMist quadrivalent vaccine may increase protection against both B strains covered by the trivalent vaccine.Citation146 Another intranasal live attenuated vaccine, Nasovac-S, has been approved in India, as its safety and efficacy were confirmed after administration to a large population.Citation147

Even though intranasal vaccines are alternatives to replace the injectable ones and have shown vaccine efficacy, there was an inactivated intranasal influenza vaccine, known as Nasalflu licensed in Switzerland, which was removed from the market because of its association with relative risk of Bell's palsy. This vaccine contained hemagglutinins from three influenza viruses (A/Bayern H1N1, B/Beijing, and A/Wuhan H3N2) and E. coli heat-labile enterotoxin (HLT). The prelicensure clinical trials of this vaccine showed no sign of Bell's palsy in 1218 vaccine recipients.Citation148,Citation149,Citation150 However, Mutsch et al. conducted a matched case-control research and a case-series analysis in German-speaking regions of Switzerland after the vaccine was licensed in Switzerland, and they found a strong association between the inactivated intranasal influenza vaccine and Bell's palsy. The disease was only observed in patients who received the intranasal vaccine but not the ones inoculated with the parenteral influenza vaccines.Citation150 They estimated that the relative risk of Bell's palsy was 19 times the risk in the controls, corresponding to 13 excess cases per 10,000 vaccinees within 1 to 91 days after vaccination. These kinds of cases have indicated the importance of preclinical safety studies, wherein it requires longer period of observation.Citation150

Transcutaneous vaccination

Transcutaneous vaccination is delivery through the skin and has several advantages, such as that the epidermis contains several types of immune cells, including DCs, langerin+ cells (LCs), T lymphocytes, NK cells, macrophages, and mast cellsCitation151,Citation152 Epidermal Langerhans cells are special DCs, responsible for controlling immune responses in the skin, and they are defined by their localization in the epidermis and the expression of CD1a and langerin (CD207). The latter molecule is a receptor of C-type lectin, which recognizes the pathogen-associated molecular pattern (PAMP) on the surface of different pathogens. After the interaction, followed by langerin-mediated endocytosis, the LC migrates to the lymph nodes to activate the immune responseCitation153,Citation154 Transcutaneous immunization may enable efficient presentation of the antigen to the immune cells of the skin, inducing a rapid and efficient immune response.

One of obstacles for transcutaneous immunization is the stratum corneum (the dead cell layer) of the skin, which is difficult to penetrate. Several studies have designed different adjuvants and delivery systems using different chemical and physical approaches to improve immune responses without the use of needles. Microneedle patches effectively deliver antigens into APCs of the epidermal and mucosal compartments. Although mucosal and systemic immunity are induced by transcutaneous immunization, the specific mechanisms are still not well understood.Citation155 Belyakov et al. demonstrated that the migration of DCs and LCs carrying an HIV peptide construct with CT, LT, or CpG oligodeoxynucleotides to the lungs induced a significant cytotoxic T lymphocyte (CTL) response in the systemic circulation and the strongest response in the lung after transcutaneous priming and transcutaneous boosting,Citation155 and these results may partially explain mucosal immunity. During this process, the LCs induced Th1 and Th2 responses after treatment with antigen-delivering microneedle vaccination. This effect was confirmed in a study in which LC-depleted mice were vaccinated with microneedles coated with subunit influenza vaccine. The results showed that 95% of the vaccine was eliminated from the skin of wild type mice, whereas 65% of the vaccine was eliminated in LC-KO mice. This mouse strain also had impaired humoral and cellular immune responses, with reduced Th1 and Th2 profiles in comparison with wild type mice.Citation156

Due to the rapid development of microfabrication manufacturing, immunization with microneedles has recently received increased attention in the vaccine field. Currently, microneedle types include solid (for direct tissue pretreatment), drug-coated, deep, dissolving, and hollow.Citation157 Several materials, including polymers, colloidal silica, ceramics, steel, glass, sugar, hydrogel, and alumina, are used for microneedle fabrication.Citation158-164 Each type of microneedle employs different mechanisms for vaccine delivery. Solid microneedles are generally made of stainless steel with a diameter of 500 μm, which directly generates skin pores and allows the penetration of topical vaccine through the surface of the pretreated skin into the body,Citation165 while drug-coated microneedles are used to release the drug slowly into the skin.Citation166 One of the materials used in drug-coated microneedles is poly(L-lactide) (PLLA), which is melted on silicone micro-molds with pyramidal cavities at 200°C, compressed by a hot press, and cooled to RT. Later, the microneedles are coated with lidocaine hydrochloride monohydrate. The investigators found that delivery efficiency into porcine skin was 69.1 ± 15.1%, 77.2 ± 13.5%, and 84.0 ± 6.8% after application at 1, 2, and 5 min, respectively, and that the lidocaine was stable for 3weeks at different temperatures.Citation166

In general, non-dissolvable microneedles are made of inorganic materials, such as metal, silicon, glass, and ceramics, while dissolvable microneedles are made from water-soluble biopolymers, such as sulfate dextran and chondroitin, in which the drugs are applied as a suspension that dissolves into the skin.Citation167 In fact, the eye may also be a target organ for microneedle application.Citation168 This technology can be applied to the delivery of DNA, viral capsid subunits, and inactivated and live attenuated vaccines. The delivery of live attenuated vaccines has several disadvantages, since the vaccine needs to be stored and distributed at low temperatures, and reconstitution is required before administration.Citation169 However, measles virus, which still greatly threatens the morbidity and mortality of children worldwide,Citation170 combined with excipient trehalose delivered by steel microneedles generates immunity against measles comparable to subcutaneous injection.Citation171 Furthermore, these microneedle measles patches are significantly protected from loss of titer for 30 days at room temperature, which meets the WHO requirements for lyophilized vaccines.Citation171,Citation172

Edens et al. further developed a dissolving microneedle patch fabricated with sucrose to evaluate the measles vaccine in rhesus macaques.Citation173 They showed that subcutaneous injection and microneedle delivery generated equivalent titers of neutralizing antibodies. However, microneedle patches sustained a higher level of activity and thermostability with increasing temperature.Citation174 In addition, Vrdoljak et al. vaccinated live recombinant adenovirus and modified vaccinia Ankara vectors by transcutaneous delivery using solid microneedle patches,Citation175 which resulted in virus delivery and infection and elicited humoral or CD8+ T cell responses comparable to that produced by intradermal injection. Similarly, an adenoviral vector vaccine delivered by microneedle conferred robust protective immunity against Zika virus and malaria.Citation176,Citation177 Motivated by the fact that the efficacy of live attenuated oral rotavirus vaccines is lower in developing countries, Wang et al. showed that the administration of inactivated rotavirus vaccine with MicronJet600® microneedles in neonatal gnotobiotic piglets generates comparable protective effects as intramuscular injection.Citation178

Muller et al. developed a high-density microprojection array, the Nanopatch, which delivers inactivated poliovirus vaccine into the skin. These microneedle arrays led to the generation of neutralizing antibodies against poliovirus in 100% of the rats treated.Citation179 Furthermore, influenza subunit vaccines were administered by microneedle patches to increase humoral immunity.Citation180,Citation181 The influenza subunit vaccine coated onto microneedle patches elicited an effective immune response comparable to intramuscular injection in guinea pigs.Citation182

Microneedles have also been used to deliver recombinant antigen vaccines, in which anthrax antigen delivered by microneedle induces the same effective immune response and protection as intramuscular injection.Citation182 Furthermore, recombinant trimeric soluble hemagglutinin derived from the A/Aichi/2/68 virus generates a higher immune response, including more antigen-specific Th1 cells, a greater mucosal antibody response, and a higher survival ratio than unmodified hemagglutinin.Citation180 Likewise, VLP vaccines confer effective protection against influenza using the microneedle route. It was shown that microneedle delivery of H5N1 influenza VLPs results in a sustained B and T cell response. Intriguingly, application to human skin induced the CD207+ Langerhans cells to migrate toward the basement membrane.Citation183 Kim et al. showed that heterologous VLPs incorporating the influenza virus M2 extracellular domain generated a broad cross-protective effect against H1N1, H3N2, and H5N1.Citation184

In another study, Hooper et al. developed an experimental smallpox DNA vaccine that included four vaccinia virus genes (4pox), which was delivered using plasmid DNA-coated microneedle arrays and induced a greater antibody response than a live virus vaccine delivered by scarification.Citation185 Moreover, vaccination with a DNA vaccine encoding influenza hemagglutinin (HA) using microneedles generated better protection against viral challenge, enhanced humoral and cellular immunity, and enabled dose sparing compared with intramuscular injection.Citation186,Citation187 Although DNA vaccines are thermostable, inexpensive, and rapidly manufactured, their use under clinical conditions is limited due to their insufficient immunogenicity.Citation188,Citation189 To solve this problem, different kinds of non-viral delivery systems, including cationic lipids, polymers, and liposomes, combined with microneedle delivery have been used for DNA vaccines.Citation190-192 Kim et al. used a pH-responsive polyelectrolyte to deliver polyplex-based DNA vaccines, which generated robust humoral and memory immune responses.Citation193 Recently, a DNA vaccine coated onto PLGA/PEI or PLGA-PLL/γPGA nanoparticles and administered with microneedle patches effectively reduced the risks of H1N1 and Ebola virus, respectively.Citation194,Citation195 Arya et al. showed that a DNA vaccine for rabies dissolved in microneedle patches generated a robust immune response, providing the potential to protect individuals from biting infection—at least to some extent.Citation196 Co-stimulation with A/PR8 influenza hemagglutinin DNA and an A/PR8 inactivated virus vaccine delivered by microneedle patches produced significant protection against A/PR8 and 2009 H1N1 virus. This study demonstrated that a single immunization with a microneedle vaccine or by intramuscular injection induced rapid memory responses with high HAI titers upon heterologous virus challenge, inducing protective immunity.Citation197

Currently, microneedle-mediated vaccine delivery has also been investigated in the clinic, and several studies have been completed (). However, the number of clinical trials of microneedle vaccines has been much fewer than the number of trials for oral or i.n. vaccines. Rouphael et al. conducted a phase I clinical trial for inactivated influenza vaccine (fluvirin) administered by microneedle. It was shown that the adverse reactogenicity is mild and transient after dissolvable microneedle administration, and patients exhibited an increased neutralizing antibody titer, seroconversion rate, and level of seroprotection similar to intramuscular injection.Citation198 Microneedle patches administered by participants themselves also generated increased immune responses.Citation198 However, no microneedle-related vaccines have been licensed to date.

Table 3. Clinical trials for transcutaneous immunization.

Further improvements

Vaccination is an important approach to controlling infectious diseases, especially in developing countries where technology and sanitary conditions are less advanced. The most common infectious diseases are malaria, TBC, HIV, and gastroenteritis produced by drinking or eating contaminated water or food. The ability to induce a balanced immune response after immunization is determined by several factors, including the route of immunization, the microorganism target organ, the nature of the antigens, the immunological vehicles, and the cytokine and T cell responses. The formulation of vaccine antigens is a problem in developing countries, where the conditions for both storage and transportation of vaccines to the countryside are suboptimal, and the cold chain may fail at either of these steps.Citation8,Citation199 Most of the vaccines are intramuscular with good results protecting millions of people, and where their administration is not needle-free, especially in developing countries where syringes are reused, it may be dangerous for both medical personnel and patients.

In this review, we have made the case that needle-free vaccines, especially those for mucosal delivery and transcutaneous immunization, are good alternatives, which induce systemic and mucosal immune responses and activate sIgA/IgG plasma B cells and T cells that pass through the lymphatic system to infection sites. However, further studies are needed to resolve some remaining issues. Currently, several clinical trials of needle-free vaccination systems are being conducted using oral, i.n., and microneedle administration, which are showing effective protection against pathogens. However, there are only a few licensed oral and i.n. vaccines that are noninvasive compared with the very large majority of vaccines that are injected. According to the clinical trial results for licensed vaccines, adverse effects may occur. Moreover, because the safety of the attenuated mucosal virus is not easy to evaluate in infants, new vaccines should be carefully evaluated before use in infants. Although there are some significant advantages of vaccination by the oral and i.n. routes, they face unique challenges. Oral vaccination can be affected by low pH, proteolytic enzymes, and biological barriers in the gastrointestinal environment.Citation3 Furthermore, the short absorption time may constrain the absorption of related vaccines.Citation200 Considering the degradation of vaccines, the vaccine dose should be increased to generate comparable effects as parenteral injection,Citation38 and this may reduce vaccine efficacy. Therefore, there are more stringent requirements for the vaccine carrier to achieve an efficient immune response, as a larger dose may lead to risks of immune tolerance, and constant antigen stimulation of the GI tract may limit the response of the GALT to biohazard antigens rather than protecting against these antigens.Citation201,Citation202

Intranasal vaccines also face short residence times and higher dose requirements.Citation101 Furthermore, physiological barriers also hinder the absorption of vaccines. Thus, solving these problems and increasing the vaccine efficacy of mucosal vaccines is still a challenge.

The other needle-free delivery system, the microneedle, has had few vaccination clinical trials. Moreover, the microneedles by themselves may not easily break the skin and may cause swelling during insertion, which has in part reduced their application in the clinic. While a recent study showed that microneedle arrays filled with liposomes loaded with hepatitis B virus vaccine induced robust systemic and mucosal immunity by oral immunization,Citation203 the alternative route of administration of microneedle arrays should also be considered. Furthermore, vaccines coated onto solid microneedles or dissolving microneedles may gradually dry out, reducing vaccine activity and possibly producing discomfort when used over a longer period. Therefore, microneedle-mediated vaccination also needs more study to improve its application in the clinic.

The mechanisms of invasive vaccines should also be elucidated for rational vaccine improvement. Overall, although noninvasive administration of vaccines still has several unsolved problems, it has a promising future, since it has the potential to reduce the cost of the vaccines in developing countries, reduce the risk of contamination with other needle-borne diseases, and speed vaccine administration during pandemics. In addition, one of the important challenges of the needle-free system is to design delivery vehicles that protect the vaccines, which should improve results in clinical applications.

Disclosure of potential conflicts of interest

The authors declare that they have no potential conflicts of interest.

Additional information

Funding

This work was supported in part by research grants AI072139 and AI133207 from the U.S. National Institute of Allergy and Infectious Diseases and the AAI Careers in Immunology Fellowship from the American Association of Immunologists (2016).

References

  • Organization WH. World health statistics 2015. Geneva (Switzerland): World Health Organization; 2015. Available online at: http://www.who.int/gho/publications/world_health_statistics/2015/en/
  • Plotkin SA. Vaccines: the fourth century. Clin Vaccine Immunol. 2009;16:1709–19. doi:10.1128/CVI.00290-09. PMID:19793898
  • Vela Ramirez JE, Sharpe LA, Peppas NA. Current state and challenges in developing oral vaccines. Adv Drug Deliv Rev. 2017;114:116–31. doi:10.1016/j.addr.2017.04.008. PMID:28438674
  • Concha C, Cañas R, Macuer J, Torres MJ, Herrada AA, Jamett F, Ibáñez C. Disease prevention: An opportunity to expand edible plant-based vaccines? Vaccines. 2017;5:14. doi:10.3390/vaccines5020014.
  • Irvine DJ, Swartz MA, Szeto GL. Engineering synthetic vaccines using cues from natural immunity. Nat Materials. 2013;12:978–90. doi:10.1038/nmat3775. PMID:24150416
  • Kim SH, Lee KY, Jang YS. Mucosal immune system and M Cell-targeting strategies for oral mucosal vaccination. Immune Network. 2012;12:165–75. doi:10.4110/in.2012.12.5.165. PMID:23213309
  • Das P. Revolutionary vaccine technology breaks the cold chain. Lancet Infect Dis. 2004;4:719. doi:10.1016/S1473-3099(04)01222-8. PMID:15593445
  • Billah MM, Zaman K, Estivariz CF, Snider CJ, Anand A, Hampton LM, Bari TIA, Russell KL, Chai SJ. Cold-chain adaptability during introduction of inactivated polio vaccine in Bangladesh, 2015. J Infect Dis. 2017;216:S114–S21. doi:10.1093/infdis/jiw591. PMID:28838173
  • Giudice EL, Campbell JD. Needle-free vaccine delivery. Adv Drug Deliv Rev. 2006;58:68–89. doi:10.1016/j.addr.2005.12.003. PMID:16564111
  • Hamilton JG. Needle phobia: a neglected diagnosis. J Fam Pract. 1995;41:169–75. PMID:7636457
  • Nir Y, Paz A, Sabo E, Potasman I. Fear of injections in young adults: Prevalence and associations. Am J Trop Med Hyg. 2003;68:341–4. PMID:12685642
  • Drucker E, Alcabes PG, Marx PA. The injection century: massive unsterile injections and the emergence of human pathogens. Lancet. 2001;358:1989–92. doi:10.1016/S0140-6736(01)06967-7. PMID:11747942
  • Kermode M. Unsafe injections in low-income country health settings: need for injection safety promotion to prevent the spread of blood-borne viruses. Health Promotion Int. 2004;19:95–103. doi:10.1093/heapro/dah110. PMID:14976177
  • Lycke N. Recent progress in mucosal vaccine development: potential and limitations. Nat Rev Immunol. 2012;12:592. doi:10.1038/nri3251. PMID:22828912
  • Fu YH, He JS, Wang XB, Zheng XX, Wu Q, Xie C, Zhang M, Wei W, Tang Q, Song JD, et al. A prime-boost vaccination strategy using attenuated Salmonella typhimurium and a replication-deficient recombinant adenovirus vector elicits protective immunity against human respiratory syncytial virus. Biochem Biophys Res Communications. 2010;395:87–92. doi:10.1016/j.bbrc.2010.03.144. PMID:20350532
  • Klavinskis LS, Barnfield C, Gao L, Parker S. Intranasal immunization with plasmid DNA-lipid complexes elicits mucosal immunity in the female genital and rectal tracts. J Immunol. 1999;162:254–62.
  • Liu L, Yao W, Rao Y, Lu X, Gao J. pH-Responsive carriers for oral drug delivery: challenges and opportunities of current platforms. Drug Deliv. 2017;24:569–81. doi:10.1080/10717544.2017.1279238. PMID:28195032
  • Seong KY, Seo MS, Hwang DY, O'Cearbhaill ED, Sreenan S, Karp JM, Yang SY. A self-adherent, bullet-shaped microneedle patch for controlled transdermal delivery of insulin. J Control Release. 2017;265:48–56. doi:10.1016/j.jconrel.2017.03.041. PMID:28344013
  • Kearney MC, Caffarel-Salvador E, Fallows SJ, McCarthy HO, Donnelly RF. Microneedle-mediated delivery of donepezil: Potential for improved treatment options in Alzheimer's disease. Eur J Pharm Biopharm. 2016;103:43–50. doi:10.1016/j.ejpb.2016.03.026. PMID:27018330
  • Hirobe S, Otsuka R, Iioka H, Quan YS, Kamiyama F, Asada H, Okada N, Nakagawa S. Clinical study of a retinoic acid-loaded microneedle patch for seborrheic keratosis or senile lentigo. Life Sci. 2017;168:24–7. doi:10.1016/j.lfs.2015.12.051. PMID:26757104
  • Hao Y, Dong ML, Zhang TY, Peng JR, Jia YP, Cao YP, Qian Z. Novel approach of using near-Infrared Responsive PEGylated Gold Nanorod Coated Poly(L-lactide) microneedles to enhance the antitumor efficiency of Docetaxel-Loaded MPEG-PDLLA Micelles for Treating an A431 tumor. Acs Appl Mater Inter. 2017;9:15317–27. doi:10.1021/acsami.7b03604.
  • Dietrich G, Griot-Wenk M, Metcalfe IC, Lang AB, Viret J-F. Experience with registered mucosal vaccines. Vaccine. 2003;21:678–83.
  • Wang S, Liu H, Zhang X, Qian F. Intranasal and oral vaccination with protein-based antigens: advantages, challenges and formulation strategies. Protein Cell. 2015;6:480–503. doi:10.1007/s13238-015-0164-2.
  • Holmgren J, Czerkinsky C. Mucosal immunity and vaccines. Nat Med. 2005;11:S45–53. doi:10.1038/nm1213. PMID:15812489
  • Strugnell RA, Wijburg OL. The role of secretory antibodies in infection immunity. Nat Rev Microbiol. 2010;8:656–67. doi:10.1038/nrmicro2384. PMID:20694027
  • Rojas R, Apodaca G. Immunoglobulin transport across polarized epithelial cells. Nat Rev Mol Cell Biol. 2002;3:944–55. doi:10.1038/nrm972. PMID:12461560
  • Shakya AK, Chowdhury MYE, Tao W, Gill HS. Mucosal vaccine delivery: Current state and a pediatric perspective. J Control Release. 2016;240:394–413. doi:10.1016/j.jconrel.2016.02.014. PMID:26860287
  • Spencer J, Finn T, Pulford K, Mason D, Isaacson P. The human gut contains a novel population of B lymphocytes which resemble marginal zone cells. Clin Exp Immunol. 1985;62:607. PMID:3910320
  • Spencer J, Sollid L. The human intestinal B-cell response. Mucosal Immunol. 2016. doi:10.1038/mi.2016.59. PMID:27461177
  • Hornquist CE, Ekman L, Grdic KD, Schon K, Lycke NY. Paradoxical IgA immunity in CD4-deficient mice. Lack of cholera toxin-specific protective immunity despite normal gut mucosal IgA differentiation. J Immunol 1995;155:2877–87. PMID:7673704
  • Fischer M, Küppers R. Human IgA‐and IgM‐secreting intestinal plasma cells carry heavily mutated VH region genes. Eur J Immunol. 1998;28:2971–7. doi:10.1002/(SICI)1521-4141(199809)28:09%3c2971::AID-IMMU2971%3e3.0.CO;2-3. PMID:9754584
  • Lindner C, Thomsen I, Wahl B, Ugur M, Sethi MK, Friedrichsen M, et al. Diversification of memory B cells drives the continuous adaptation of secretory antibodies to gut microbiota. Nat Immunol. 2015;16:880–8. doi:10.1038/ni.3213. PMID:26147688
  • Bernstein JM, Baekkevold ES, Brandtzaeg P. Immunobiology of the Tonsils and Adenoids-Chapter 90.
  • Kunkel EJ, Butcher EC. Plasma-cell homing. Nat Rev Immunol. 2003;3:822–9. doi:10.1038/nri1203. PMID:14523388
  • Hunter MC, Teijeira A, Halin C. T cell trafficking through lymphatic vessels. Frontiers Immunol. 2016;7:613. doi:10.3389/fimmu.2016.00613.
  • Staats HF, Jackson RJ, Marinaro M, Takahashi I, Kiyono H, McGhee JR. Mucosal immunity to infection with implications for vaccine development. Curr Opin Immunol. 1994;6:572–83. doi:10.1016/0952-7915(94)90144-9. PMID:7946045
  • Marasini N, Skwarczynski M, Toth I. Oral delivery of nanoparticle-based vaccines. Exp Rev Vaccin. 2014;13:1361–76. doi:10.1586/14760584.2014.936852. PMID:25155636
  • Pavot V, Rochereau N, Genin C, Verrier B, Paul S. New insights in mucosal vaccine development. Vaccine. 2012;30:142–54. doi:10.1016/j.vaccine.2011.11.003. PMID:22085556
  • Schulz O, Pabst O. Antigen sampling in the small intestine. Trends Immunol. 2013;34:155–61. doi:10.1016/j.it.2012.09.006. PMID:23083727
  • Lee EY, Kim JY, Lee DK, Yoon IS, Ko HL, Chung JW, Chang J, Nam JH. Sublingual immunization with Japanese encephalitis virus vaccine effectively induces immunity through both cellular and humoral immune responses in mice. Microbiol Immunol. 2016;60:846–53. doi:10.1111/1348-0421.12458. PMID:28004418
  • Kamble NM, Hajam IA, Lee JH. Orally administered live attenuated Salmonella Typhimurium protects Q CrossMark mice against lethal infection with H1N1 influenza virus. Vet Microbiol. 2017;201:1–6. doi:10.1016/j.vetmic.2017.01.006. PMID:28284594
  • Wang L, Wang XH, Bi K, Sun XM, Yang J, Gu Y, et al. Oral vaccination with Attenuated Salmonella typhimurium-Delivered TsPmy DNA vaccine Elicits protective immunity against Trichinella spiralis in BALB/c Mice. Plos Neglect Trop D. 2016;10(9): e0004952. doi:10.1371/journal.pntd.0004952.
  • Lubeck MD, Davis AR, Chengalvala M, Natuk RJ, Morin JE, Molnar-Kimber K, Mason BB, Bhat BM, Mizutani S, Hung PP, et al. Immunogenicity and efficacy testing in chimpanzees of an oral hepatitis B vaccine based on live recombinant adenovirus. Proc Natl Acad Sci U S A. 1989;86:6763–7. doi:10.1073/pnas.86.17.6763. PMID:2570422
  • Charlton KM, Artois M, Prevec L, Campbell JB, Casey GA, Wandeler AI, Armstrong J. Oral rabies vaccination of skunks and foxes with a recombinant human adenovirus vaccine. Arch Virol. 1992;123:169–79. doi:10.1007/BF01317147. PMID:1550495
  • Fooks AR, Jeevarajah D, Lee J, Warnes A, Niewiesk S, ter Meulen V, Stephenson JR, Clegg JC. Oral or parenteral administration of replication-deficient adenoviruses expressing the measles virus haemagglutinin and fusion proteins: protective immune responses in rodents. J General Virol. 1998;79 (Pt 5):1027–31. doi:10.1099/0022-1317-79-5-1027. PMID:9603317
  • Natuk RJ, Lubeck MD, Chanda PK, Chengalvala M, Wade MS, Murthy SC, Wilhelm J, Vernon SK, Dheer SK, Mizutani S, et al. Immunogenicity of recombinant human adenovirus-human immunodeficiency virus vaccines in chimpanzees. AIDS Res Hum Retroviruses. 1993;9:395–404. doi:10.1089/aid.1993.9.395. PMID:8318268
  • Patel A, Zhang Y, Croyle M, Tran K, Gray M, Strong J, Feldmann H, Wilson JM, Kobinger GP. Mucosal delivery of adenovirus-based vaccine protects against Ebola virus infection in mice. J Infect Dis. 2007;196 Suppl 2:S413–20. doi:10.1086/520603. PMID:17940978
  • Chen S, Xu Q, Zeng M. Oral vaccination with an adenovirus-vectored vaccine protects against botulism. Vaccine. 2013;31:1009–11. doi:10.1016/j.vaccine.2012.12.054. PMID:23295065
  • Gurwith M, Lock M, Taylor EM, Ishioka G, Alexander J, Mayall T, Ervin JE, Greenberg RN, Strout C, Treanor JJ, et al. Safety and immunogenicity of an oral, replicating adenovirus serotype 4 vector vaccine for H5N1 influenza: a randomised, double-blind, placebo-controlled, phase 1 study. Lancet Infect Dis. 2013;13:238–50. doi:10.1016/S1473-3099(12)70345-6.
  • Lin J, Tu C, Mou C, Chen X, Yang Q. CpG DNA facilitate the inactivated transmissible gastroenteritis virus in enhancing the local and systemic immune response of pigs via oral administration. Veterinary Immunol Immunopathol. 2016;172:1–8. doi:10.1016/j.vetimm.2016.02.013.
  • Edwards M, Dimmock N. Two influenza A virus-specific Fabs neutralize by inhibiting virus attachment to target cells, while neutralization by their IgGs is complex and occurs simultaneously through fusion inhibition and attachment inhibition. Virology. 2000;278:423–35. doi:10.1006/viro.2000.0631. PMID:11118365
  • Li W, Shi W, Qiao H, Ho SY, Luo A, Zhang Y, Zhu C. Positive selection on hemagglutinin and neuraminidase genes of H1N1 influenza viruses. Virol J. 2011;8:183. doi:10.1186/1743-422X-8-183. PMID:21507270
  • Brandtzaeg P. Mucosal immunity: induction, dissemination, and effector functions. Scandinavian J immunol. 2009;70:505–15. doi:10.1111/j.1365-3083.2009.02319.x. PMID:19906191
  • Shim B-S, Choi Y, Cheon IS, Song MK. Sublingual delivery of vaccines for the induction of mucosal immunity. Immune Network 2013;13:81–5. doi:10.4110/in.2013.13.3.81. PMID:23885221
  • Spinner JL, Oberoi HS, Yorgensen YM, Poirier DS, Burkhart DJ, Plante M, Evans JT. Methylglycol chitosan and a synthetic TLR4 agonist enhance immune responses to influenza vaccine administered sublingually. Vaccine. 2015;33:5845–53. doi:10.1016/j.vaccine.2015.08.086. PMID:26392012
  • Conner ME, Crawford SE, Barone C, O'Neal C, Zhou YJ, Fernandez F, Parwani A, Saif LJ, Cohen J, Estes MK. Rotavirus subunit vaccines. Arch Virol Suppl. 1996;12:199–206. PMID:9015116
  • Zhou ZW, Dong H, Huang YM, Yao SW, Liang BS, Xie YQ, Long Y, Mai J, Gong S. Recombinant Bacillus subtilis spores expressing cholera toxin B subunit and Helicobacter pylori urease B confer protection against H. pylori in mice. J Med Microbiol. 2017;66:83–9. doi:10.1099/jmm.0.000404. PMID:27930268
  • Borde A, Ekman A, Larsson A, Carlin N, Holmgren J, Tobias J. Preparation and preclinical evaluation of a freeze-dried formulation of a novel combined multivalent whole-cell/B-subunit oral vaccine against enterotoxigenic Escherichia coli diarrhea. Eur J Pharmaceutics Biopharmaceutics 2016;108:18–24. doi:10.1016/j.ejpb.2016.07.011. PMID:27485398
  • Yagnik B, Sharma D, Padh H, Desai P. Immunization with r-Lactococcus lactis expressing outer membrane protein A of Shigella dysenteriae type-1: evaluation of oral and intranasal route of administration. J Applied Microbiol. 2017;122:493–505. doi:10.1111/jam.13353. PMID:27860045
  • Oliveira CR, Rezende CMF, Silva MR, Borges OM, Pego AP, Goes AM. Oral vaccination based on DNA-Chitosan Nanoparticles against Schistosoma mansoni infection. Sci World J. 2012;2012:938457. doi:10.1100/2012/938457.
  • Jazayeri SD, Ideris A, Zakaria Z, Shameli K, Moeini H, Omar AR. Cytotoxicity and immunological responses following oral vaccination of nanoencapsulated avian influenza virus H5 DNA vaccine with green synthesis silver nanoparticles. J Controlled Release. 2012;161:116–23. doi:10.1016/j.jconrel.2012.04.015. PMID:22549012
  • Baldauf KJ, Royal JM, Kouokam JC, Haribabu B, Jala VR, Yaddanapudi K, Hamorsky KT, Dryden GW, Matoba N. Oral administration of a recombinant cholera toxin B subunit promotes mucosal healing in the colon. Mucosal Immunol. 2017;10:887–900. doi:10.1038/mi.2016.95. PMID:27805617
  • Tarakanovskaya MG, Chinburen J, Batchuluun P, Munkhzaya C, Purevsuren G, Dandii D, Hulan T, Oyungerel D, Kutsyna GA, Reid AA, et al. Open-label Phase II clinical trial in 75 patients with advanced hepatocellular carcinoma receiving daily dose of tableted liver cancer vaccine, hepcortespenlisimut-L. J Hepatocell Carcino. 2017;4:59–69. doi:10.2147/JHC.S122507.
  • Azegami T, Itoh H, Kiyono H, Yuki Y. Novel transgenic rice-based vaccines. Archivum Immunologiae Et Therapiae Experimentalis 2015;63:87–99. doi:10.1007/s00005-014-0303-0. PMID:25027548
  • Fukuda K, Ishida W, Harada Y, Wakasa Y, Takagi H, Takaiwa F, et al. Efficacy of oral immunotherapy with a rice-based edible vaccine containing hypoallergenic Japanese cedar pollen allergens for treatment of established allergic conjunctivitis in mice. Allergol Int. 2018;67(1):119–23. doi: 10.1016/j.alit.2017.06.006. PMID: 28676431.
  • Ogawa T, Kashima K, Yuki Y, Mejima M, Kurokawa S, Kuroda M, Okazawa A, Kiyono H, Ohta D. Seed metabolome analysis of a transgenic rice line expressing cholera Toxin B-subunit. Scientific Reports. 2017;7:5196. doi:10.1038/s41598-017-04701-w. PMID:28701756
  • Takeyama N, Yuki Y, Tokuhara D, Oroku K, Mejima M, Kurokawa S, Kuroda M, Kodama T, Nagai S, Ueda S, et al. Oral rice-based vaccine induces passive and active immunity against enterotoxigenic E. coli-mediated diarrhea in pigs. Vaccine. 2015;33:5204–11. doi:10.1016/j.vaccine.2015.07.074. PMID:26254309
  • Changotra H, Vij A. Rotavirus virus‐like particles (RV‐VLPs) vaccines: An update. Rev Medical Virol. 2017;27(6):e1954. doi:10.1002/rmv.1954. PMID:29048711
  • Dang DA, Nguyen VT, Vu DT, Nguyen TH, Nguyen DM, Yuhuan W, Baoming J, Nguyen DH, Le TL. A dose-escalation safety and immunogenicity study of a new live attenuated human rotavirus vaccine (Rotavin-M1) in Vietnamese children. Vaccine. 2012;30 Suppl 1:A114–21. PMID:22520120
  • Chen WH, Garza J, Choquette M, Hawkins J, Hoeper A, Bernstein DI, Cohen MB. Safety and immunogenicity of escalating dosages of a single oral administration of peru-15 pCTB, a candidate live, attenuated vaccine against enterotoxigenic Escherichia coli and Vibrio cholerae. Clin Vaccine Immunol. 2015;22:129–35. doi:10.1128/CVI.00560-14. PMID:25410205
  • Liebowitz D, Lindbloom JD, Brandl JR, Garg SJ, Tucker SN. High titre neutralising antibodies to influenza after oral tablet immunisation: a phase 1, randomised, placebo-controlled trial. Lancet Infect Dis. 2015;15:1041–8. doi:10.1016/S1473-3099(15)00266-2. PMID:26333337
  • Baik YO, Choi SK, Kim JW, Yang JS, Kim IY, Kim CW, Hong JH. Safety and immunogenicity assessment of an oral cholera vaccine through phase I clinical trial in Korea. J Korean Medical Sci. 2014;29:494–501. doi:10.3346/jkms.2014.29.4.494. PMID:24753695
  • Zeng M, Mao XH, Li JX, Tong WD, Wang B, Zhang YJ, Guo G, Zhao ZJ, Li L, Wu DL, et al. Efficacy, safety, and immunogenicity of an oral recombinant Helicobacter pylori vaccine in children in China: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2015;386:1457–64. doi:10.1016/S0140-6736(15)60310-5. PMID:26142048
  • Peters W, Brandl JR, Lindbloom JD, Martinez CJ, Scallan CD, Trager GR, Tingley DW, Kabongo ML, Tucker SN. Oral administration of an adenovirus vector encoding both an avian influenza A hemagglutinin and a TLR3 ligand induces antigen specific granzyme B and IFN-gamma T cell responses in humans. Vaccine. 2013;31:1752–8. doi:10.1016/j.vaccine.2013.01.023. PMID:23357198
  • Hamborsky J, Kroger A, Wolfe S, Control CfD, Prevention. Epidemiology and prevention of vaccine-preventable diseases. US Dep Health Hum Services, Centers Dis Control Prevention. 2015. Available online at: https://www.cdc.gov/vaccines/pubs/pinkbook/index.html
  • Hammon WM, Coriell LL, Ludwig EH, McAllister RM, Greene AE, Sather GE, Wehrle PF. Evaluation of Red Cross gamma globulin as a prophylactic agent for poliomyelitis: 5. Reanalysis of results based on laboratory-confirmed cases. J Am Medical Assoc. 1954;156:21–7. doi:10.1001/jama.1954.02950010023009. PMID:13183798
  • Koprowski H, Jervis GA, Norton TW. Immune responses in human volunteers upon oral administration of a rodent-adapted strain of poliomyelitis virus. Am J Epidemiol. 1952;55:108–26. doi:10.1093/oxfordjournals.aje.a119499.
  • Salk JE, Bazeley P, Bennett BL, Krech U, Lewis LJ, Ward EN, et al. Ii. a practical means for inducing and maintaining antibody formation. Am J Public Health Nations Health. 1954;44:994–1009. doi:10.2105/AJPH.44.8.994.
  • Kew OM, Sutter RW, de Gourville EM, Dowdle WR, Pallansch MA. Vaccine-derived polioviruses and the endgame strategy for global polio eradication. Annu Rev Microbiol 2005;59:587–635. doi:10.1146/annurev.micro.58.030603.123625. PMID:16153180
  • Sabin AB. Oral poliovirus vaccine: history of its development and use and current challenge to eliminate poliomyelitis from the world. The University of Chicago Press. J Infect Dis. 1985;151(3):420–36. doi:10.1093/infdis/151.3.420. PMID:2982959
  • Platt LR, Estívariz CF, Sutter RW. Vaccine-associated paralytic poliomyelitis: a review of the epidemiology and estimation of the global burden. J Infect Dis. 2014;210:S380–S9. doi:10.1093/infdis/jiu184. PMID:25316859
  • WHO. Poliomyelitis (Polio), accessed on February 21, 2018. http://www.who.int/ith/vaccines/polio/en/
  • Top FH, Jr. Control of adenovirus acute respiratory disease in U.S. Army trainees. Yale J Biol Med. 1975;48:185–95. PMID:1099823
  • Kuschner RA, Russell KL, Abuja M, Bauer KM, Faix DJ, Hait H, Henrick J, Jacobs M, Liss A, Lynch JA, et al. A phase 3, randomized, double-blind, placebo-controlled study of the safety and efficacy of the live, oral adenovirus type 4 and type 7 vaccine, in U.S. military recruits. Vaccine. 2013;31:2963–71. doi:10.1016/j.vaccine.2013.04.035. PMID:23623865
  • Mosley JF, 2nd, Smith LL, Brantley P, Locke D, Como M. Vaxchora: The First FDA-Approved Cholera Vaccination in the United States. P & T. 2017;42:638–40.
  • Buyse H, Vinals C, Karkada N, Han HH. The human rotavirus vaccine Rotarix in infants: an integrated analysis of safety and reactogenicity. Hum Vaccin Immunother. 2014;10:19–24. doi:10.4161/hv.26476.
  • Justino MC, Araujo EC, van Doorn LJ, Oliveira CS, Gabbay YB, Mascarenhas JD, Miranda YS, Guerra Sde F, Silva VB, Linhares AC. Oral live attenuated human rotavirus vaccine (Rotarix) offers sustained high protection against severe G9P[8]rotavirus gastroenteritis during the first two years of life in Brazilian children. Mem Inst Oswaldo Cruz. 2012;107:846–53. doi:10.1590/S0074-02762012000700002. PMID:23147138
  • Armah GE, Breiman RF, Tapia MD, Dallas MJ, Neuzil KM, Binka FN, Sow SO, Ojwando J, Ciarlet M, Steele AD. Immunogenicity of the pentavalent rotavirus vaccine in African infants. Vaccine. 2012;30 Suppl 1:A86–93. doi:10.1016/j.vaccine.2011.10.006. PMID:22520142
  • Ward RL, Bernstein DI, Smith VE, Sander DS, Shaw A, Eiden JJ, Heaton P, Offit PA, Clark HF. Rotavirus immunoglobulin a responses stimulated by each of 3 doses of a quadrivalent human/bovine reassortant rotavirus vaccine. J Infect Dis. 2004;189:2290–3. doi:10.1086/421248. PMID:15181577
  • Vesikari T. Rotavirus vaccination: a concise review. Clin Microbiol Infect. 2012;18 Suppl 5:57–63. doi:10.1111/j.1469-0691.2012.03981.x. PMID:22882248
  • de Jong HK, Garcia-Laorden MI, Hoogendijk AJ, Parry CM, Maude RR, Dondorp AM, Faiz MA, van der Poll T, Wiersinga WJ. Expression of intra- and extracellular granzymes in patients with typhoid fever. PLoS Negl Trop Dis. 2017;11:e0005823. doi:10.1371/journal.pntd.0005823. PMID:28749963
  • Nisini R, Biselli R, Matricardi PM, Fattorossi A, D'Amelio R. Clinical and immunological response to typhoid vaccination with parenteral or oral vaccines in two groups of 30 recruits. Vaccine. 1993;11:582–6. doi:10.1016/0264-410X(93)90237-R. PMID:8488716
  • Salerno-Goncalves R, Pasetti MF, Sztein MB. Characterization of CD8(+) effector T cell responses in volunteers immunized with Salmonella enterica serovar Typhi strain Ty21a typhoid vaccine. J Immunol. 2002;169:2196–203. doi:10.4049/jimmunol.169.4.2196. PMID:12165550
  • Lundin BS, Johansson C, Svennerholm AM. Oral immunization with a Salmonella enterica serovar typhi vaccine induces specific circulating mucosa-homing CD4(+) and CD8(+) T cells in humans. Infect Immun. 2002;70:5622–7. doi:10.1128/IAI.70.10.5622-5627.2002. PMID:12228290
  • Saha A, Chowdhury MI, Khanam F, Bhuiyan MS, Chowdhury F, Khan AI, Khan IA, Clemens J, Ali M, Cravioto A, et al. Safety and immunogenicity study of a killed bivalent (O1 and O139) whole-cell oral cholera vaccine Shanchol, in Bangladeshi adults and children as young as 1 year of age. Vaccine. 2011;29:8285–92. doi:10.1016/j.vaccine.2011.08.108. PMID:21907255
  • Begue RE, Castellares G, Ruiz R, Hayashi KE, Sanchez JL, Gotuzzo E, Oberst RB, Taylor DN, Svennerholm AM. Community-based assessment of safety and immunogenicity of the whole cell plus recombinant B subunit (WC/rBS) oral cholera vaccine in Peru. Vaccine. 1995;13:691–4. doi:10.1016/0264-410X(94)00056-S. PMID:7668039
  • Sanchez JL, Trofa AF, Taylor DN, Kuschner RA, DeFraites RF, Craig SC, Rao MR, Clemens JD, Svennerholm AM, Sadoff JC, et al. Safety and immunogenicity of the oral, whole cell/recombinant B subunit cholera vaccine in North American volunteers. J Infect Dis. 1993;167:1446–9. doi:10.1093/infdis/167.6.1446. PMID:8501336
  • Sirskyj D, Kumar A, Azizi A. Mechanisms underlying the immune response generated by an oral vibrio cholerae vaccine. Int J Mol Sci. 2016;17(7): E1062. doi:10.3390/ijms17071062. PMID: 27384558
  • Alam MM, Riyadh MA, Fatema K, Rahman MA, Akhtar N, Ahmed T, Chowdhury MI, Chowdhury F, Calderwood SB, Harris JB, et al. Antigen-specific memory B-cell responses in Bangladeshi adults after one- or two-dose oral killed cholera vaccination and comparison with responses in patients with naturally acquired cholera. Clin Vaccine Immunol. 2011;18:844–50. doi:10.1128/CVI.00562-10. PMID:21346055
  • Pires A, Fortuna A, Alves G, Falcao A. Intranasal drug delivery: how, why and what for? J Pharm Pharm Sci. 2009;12:288–311. doi:10.18433/J3NC79.
  • Riese P, Sakthivel P, Trittel S, Guzman CA. Intranasal formulations: promising strategy to deliver vaccines. Exp Opin Drug Delivery. 2014;11:1619–34. doi:10.1517/17425247.2014.931936. PMID:24962722
  • Fan SF, Gao YW, Shinya K, Li CK, Li YB, Shi JZ, Jiang Y, Suo Y, Tong T, Zhong G, et al. Immunogenicity and protective efficacy of a live Attenuated H5N1 vaccine in nonhuman primates. PLoS Pathogens. 2009;5:e1000409. doi:10.1371/journal.ppat.1000409. PMID:19412338
  • Yang PH, Duan YQ, Wang C, Xing L, Gao XA, Tang C, Luo D, Zhao Z, Jia W, Peng D, et al. Immunogenicity and protective efficacy of a live attenuated vaccine against the 2009 pandemic A H1N1 in mice and ferrets. Vaccine. 2011;29:698–705. doi:10.1016/j.vaccine.2010.11.026. PMID:21111782
  • Dwivedi V, Manickam C, Patterson R, Dodson K, Murtaugh M, Torrelles JB, Schlesinger LS, Renukaradhya GJ. Cross-protective immunity to porcine reproductive and respiratory syndrome virus by intranasal delivery of a live virus vaccine with a potent adjuvant. Vaccine. 2011;29:4058–66. doi:10.1016/j.vaccine.2011.03.006. PMID:21419162
  • Li JW, Diaz-Arevalo D, Chen YP, Zeng MT. Intranasal vaccination with an engineered influenza virus expressing the receptor binding subdomain of botulinum neurotoxin provides protective immunity against botulism and influenza. Frontiers Immunol. 2015;6:170. doi:10.3389/fimmu.2015.00170. PMID: 25954272
  • Li JW, Arevalo MT, Diaz-Arevalo D, Chen YP, Choi JG, Zeng MT. Generation of a safe and effective live viral vaccine by virus self-attenuation using species-specific artificial microRNA. J Controlled Release. 2015;207:70–6. doi:10.1016/j.jconrel.2015.04.001. PMID:25858415
  • Huang MH, Dai SH, Chong PL. Mucosal delivery of a combination adjuvant comprising emulsified fine particles and LD-indolicidin enhances serological immunity to inactivated influenza virus. Microbes Infect. 2016;18:706–9. doi:10.1016/j.micinf.2016.06.007. PMID:27394146
  • Wong G, Richardson JS, Cutts T, Qiu XG, Kobinger GP. Intranasal immunization with an adenovirus vaccine protects guinea pigs from Ebola virus transmission by infected animals. Antiviral Res. 2015;116:17–9. doi:10.1016/j.antiviral.2015.01.001. PMID:25596432
  • Malley R, Lipsitch M, Stack A, Saladino R, Fleisher G, Pelton S, Thompson C, Briles D, Anderson P. Intranasal immunization with killed unencapsulated whole cells prevents colonization and invasive disease by capsulated pneumococci. Infect Immunity. 2001;69:4870–3. doi:10.1128/IAI.69.8.4870-4873.2001. PMID:11447162
  • Okamoto S, Kawabata S, Fujitaka H, Uehira T, Okuno Y, Hamada S. Vaccination with formalin-inactivated influenza vaccine protects mice against lethal influenza Streptococcus pyogenes superinfection. Vaccine. 2004;22:2887–93. doi:10.1016/j.vaccine.2003.12.024. PMID:15246625
  • Kraan H, Soema P, Amorij JP, Kersten G. Intranasal and sublingual delivery of inactivated polio vaccine. Vaccine. 2017;35:2647–53. doi:10.1016/j.vaccine.2017.03.090. PMID:28400164
  • Harakuni T, Kohama H, Tadano M, Uechi G, Tsuji N, Matsumoto Y, Miyata T, Tsuboi T, Oku H, Arakawa T. Mucosal Vaccination Approach against Mosquito-Borne Japanese Encephalitis Virus. Jpn J Infect Dis. 2009;62:37–45. PMID:19168957
  • Arnaboldi PM, Sambir M, D'Arco C, Peters LA, Seegers JFML, Mayer L, McCormick AA, Dattwyler RJ. Intranasal delivery of a protein subunit vaccine using a Tobacco Mosaic Virus platform protects against pneumonic plague. Vaccine. 2016;34:5768–76. doi:10.1016/j.vaccine.2016.09.063. PMID:27745954
  • Zeng WG, Tan ACL, Horrocks K, Jackson DC. A lipidated form of the extracellular domain of influenza M2 protein as a self-adjuvanting vaccine candidate. Vaccine. 2015;33:3526–32. doi:10.1016/j.vaccine.2015.05.053. PMID:26049002
  • Nochi T, Yuki Y, Takahashi H, Sawada S, Mejima M, Kohda T, Harada N, Kong IG, Sato A, Kataoka N, et al. Nanogel antigenic protein-delivery system for adjuvant-free intranasal vaccines. Nat Mater. 2010;9:572–8. doi:10.1038/nmat2784. PMID:20562880
  • Li JW, Arevalo MT, Chen YP, Posadas O, Smith JA, Zeng MT. Intranasal immunization with influenza antigens conjugated with cholera toxin subunit B stimulates broad spectrum immunity against influenza viruses. Hum Vaccin Immunother. 2014;10:1211–20. doi:10.4161/hv.28407.
  • Sakai S, Takashima Y, Matsumoto Y, Reed SG, Hayashi Y, Matsumoto Y. Intranasal immunization with Leish-111f induces IFN-gamma production and protects mice from Leishmania major infection. Vaccine. 2010;28:2207–13. doi:10.1016/j.vaccine.2009.12.055. PMID:20056184
  • Cui YL, Zhang XM, Gong Y, Niu SQ, Yin NL, Yao R, Xu W, Li D, Wang H, He Y, et al. Immunization with DnaJ (hsp40) could elicit protection against nasopharyngeal colonization and invasive infection caused by different strains of Streptococcus pneumoniae. Vaccine. 2011;29:1736–44. doi:10.1016/j.vaccine.2010.12.126. PMID:21238570
  • Medina M, Villena J, Salva S, Vintini E, Langella P, Alvarez S. Nasal administration of Lactococcus lactis improves local and systemic immune responses against Streptococcus pneumoniae. Microbiol Immunol. 2008;52:399–409. doi:10.1111/j.1348-0421.2008.00050.x. PMID:18667039
  • Maunder HE, Taylor G, Leppard KN, Easton AJ. Intranasal immunisation with recombinant adenovirus vaccines protects against a lethal challenge with pneumonia virus of mice. Vaccine. 2015;33:6641–9. doi:10.1016/j.vaccine.2015.10.105. PMID:26529077
  • Rivera-Hernandez T, Hartas J, Wu Y, Chuan YP, Lua LHL, Good M, Batzloff MR, Middelberg AP. Self-adjuvanting modular virus-like particles for mucosal vaccination against group A streptococcus (GAS). Vaccine. 2013;31:1950–5. doi:10.1016/j.vaccine.2013.02.013. PMID:23422147
  • Cai MY, Wang C, Li YF, Gu HJ, Sun SJ, Duan YQ, et al. Virus-like particle vaccine by intranasal vaccination elicits protective immunity against respiratory syncytial viral infection in mice. Acta Bioch Bioph Sin. 2017;49:74–82. doi:10.1093/abbs/gmw118.
  • Poteet E, Lewis P, Chen CY, Ho SO, Do T, Chiang SM, Labranche C, Montefiori D, Fujii G, Yao Q. Toll-like receptor 3 adjuvant in combination with virus-like particles elicit a humoral response against HIV. Vaccine. 2016;34:5886–94. doi:10.1016/j.vaccine.2016.10.036. PMID:27997339
  • Ji XL, Ren ZG, Xu N, Meng LN, Yu ZJ, Feng N, Sang X, Li S, Li Y, Wang T, et al. Intranasal immunization with influenza virus-like particles containing membrane-anchored Cholera Toxin B or Ricin Toxin B enhances adaptive immune responses and protection against an antigenically distinct virus. Viruses-Basel. 2016;8:115. doi:10.3390/v8040115.
  • Hjelm BE, Kilbourne J, Herbst-Kralovetz MM. TLR7 and 9 agonists are highly effective mucosal adjuvants for norovirus virus-like particle vaccines. Hum Vaccin Immunother. 2014;10:410–6. doi:10.4161/hv.27147.
  • Jackson EM, Herbst-Kralovetz MM. Intranasal Vaccination with Murabutide Enhances Humoral and Mucosal immune responses to a virus-like particle vaccine. Plos One. 2012;7:e41529. doi:10.1371/journal.pone.0041529. PMID: 22855691
  • Riquelme-Neira R, Rivera A, Saez D, Fernandez P, Osorio G, del Canto F, et al. Vaccination with DNA Encoding Truncated Enterohemorrhagic Escherichia coli (EHEC) Factor for Adherence-1 Gene (efa-1′) Confers Protective Immunity to Mice Infected with E. coli O157:H7. Front Cell Infect Microbiol. 2015;5:104. PMID:26835434
  • Su LK, Yu F, Li ZF, Zeng C, Xu QA, Fan MW. Intranasal co-delivery of IL-6 gene enhances the immunogenicity of anti-caries DNA vaccine. Acta Pharmacologica Sinica. 2014;35:592–8. doi:10.1038/aps.2013.184. PMID:24705100
  • Rolland-Debord C, Lair D, Roussey-Bihouee T, Hassoun D, Evrard J, Cheminant MA, Chesné J, Braza F, Mahay G, Portero V, et al. Block copolymer/DNA vaccination induces a strong allergen-specific local response in a mouse model of house dust mite asthma. PLoS One. 2014;9:e85976. doi:10.1371/journal.pone.0085976. PMID:24497934
  • Qin T, Yin Y, Huang L, Yu Q, Yang Q. H9N2 influenza whole inactivated virus combined with polyethyleneimine strongly enhances mucosal and systemic immunity after intranasal immunization in mice. Clin Vaccine Immunol. 2015;22:421–9. doi:10.1128/CVI.00778-14. PMID:25673304
  • Zhao K, Rong G, Hao Y, Yu L, Kang H, Wang X, Wang X, Jin Z, Ren Z, Li Z. IgA response and protection following nasal vaccination of chickens with Newcastle disease virus DNA vaccine nanoencapsulated with Ag@SiO2 hollow nanoparticles. Sci Rep. 2016;6:25720. doi:10.1038/srep25720. PMID:27170532
  • Li M, Zhao MN, Fu Y, Li Y, Gong T, Zhang ZR, Sun X. Enhanced intranasal delivery of mRNA vaccine by overcoming the nasal epithelial barrier via intra- and paracellular pathways. J Controlled Release. 2016;228:9–19. doi:10.1016/j.jconrel.2016.02.043. PMID:26941035
  • Gomez M, Mufson MA, Dubovsky F, Knightly C, Zeng W, Losonsky G. Phase-I study MEDI-534, of a live, attenuated intranasal vaccine against respiratory syncytial virus and parainfluenza-3 virus in seropositive children. Pediatric Infect Dis J. 2009;28:655–8. doi:10.1097/INF.0b013e318199c3b1. PMID:19483659
  • Malkin E, Yogev R, Abughali N, Sliman J, Wang CK, Zuo F, Yang CF, Eickhoff M, Esser MT, Tang RS, et al. Safety and immunogenicity of a live attenuated RSV vaccine in healthy RSV-seronegative children 5 to 24 months of age. PLoS One. 2013;8:e77104. doi:10.1371/journal.pone.0077104. PMID:24204744
  • Bernstein DI, Malkin E, Abughali N, Falloon J, Yi T, Dubovsky F, MI-CP149 Investigators. Phase 1 study of the safety and immunogenicity of a live, attenuated respiratory syncytial virus and parainfluenza virus type 3 vaccine in seronegative children. Pediatric Infect Dis J. 2012;31:109–14. doi:10.1097/INF.0b013e31823386f1.
  • Yang CF, Wang CK, Malkin E, Schickli JH, Shambaugh C, Zuo F, Galinski MS, Dubovsky F, Study Group, Tang RS. Implication of respiratory syncytial virus (RSV) F transgene sequence heterogeneity observed in Phase 1 evaluation of MEDI-534, a live attenuated parainfluenza type 3 vectored RSV vaccine. Vaccine. 2013;31:2822–7. doi:10.1016/j.vaccine.2013.04.006. PMID:23602668
  • Thorstensson R, Trollfors B, Al-Tawil N, Jahnmatz M, Bergstrom J, Ljungman M, Bergström J, Ljungman M, Törner A, Wehlin L, et al. A phase I clinical study of a live attenuated Bordetella pertussis vaccine–BPZE1; a single centre, double-blind, placebo-controlled, dose-escalating study of BPZE1 given intranasally to healthy adult male volunteers. PLoS One. 2014;9:e83449. doi:10.1371/journal.pone.0083449. PMID:24421886
  • Jahnmatz M, Amu S, Ljungman M, Wehlin L, Chiodi F, Mielcarek N, Locht C, Thorstensson R. B-cell responses after intranasal vaccination with the novel attenuated Bordetella pertussis vaccine strain BPZE1 in a randomized phase I clinical trial. Vaccine. 2014;32:3350–6. doi:10.1016/j.vaccine.2014.04.048. PMID:24793938
  • Schnoeller C, Roux X, Sawant D, Raze D, Olszewska W, Locht C, Openshaw PJ. Attenuated Bordetella pertussis vaccine protects against respiratory syncytial virus disease via an IL-17-dependent mechanism. Am J Respir Critical Care Med. 2014;189:194–202.
  • Brekke K, Lind A, Holm-Hansen C, Haugen IL, Sorensen B, Sommerfelt M, Kvale D. Intranasal administration of a therapeutic HIV vaccine (Vacc-4x) induces dose-dependent systemic and mucosal immune responses in a randomized controlled trial. PLoS One. 2014;9:e112556. doi:10.1371/journal.pone.0112556. PMID:25398137
  • Atmar RL, Bernstein DI, Harro CD, Al-Ibrahim MS, Chen WH, Ferreira J, Estes MK, Graham DY, Opekun AR, Richardson C, et al. Norovirus vaccine against experimental human Norwalk Virus illness. N Engl J Med. 2011;365:2178–87. doi:10.1056/NEJMoa1101245. PMID:22150036
  • El-Kamary SS, Pasetti MF, Mendelman PM, Frey SE, Bernstein DI, Treanor JJ, Ferreira J, Chen WH, Sublett R, Richardson C, et al. Adjuvanted intranasal Norwalk virus-like particle vaccine elicits antibodies and antibody-secreting cells that express homing receptors for mucosal and peripheral lymphoid tissues. J Infect Dis. 2010;202:1649–58. doi:10.1086/657087. PMID:20979455
  • Lambkin-Williams R, Gelder C, Broughton R, Mallett CP, Gilbert AS, Mann A, He D, Oxford JS, Burt D. An intranasal proteosome-adjuvanted trivalent influenza vaccine is safe, immunogenic & efficacious in the human viral influenza challenge model. Serum IgG & Mucosal IgA are important correlates of protection against illness associated with infection. PLoS One. 2016;11:e0163089. doi:10.1371/journal.pone.0163089. PMID:28005959
  • Mallory RM, Malkin E, Ambrose CS, Bellamy T, Shi L, Yi T, Jones T, Kemble G, Dubovsky F. Safety and immunogenicity following administration of a live, attenuated monovalent 2009 H1N1 influenza vaccine to children and adults in two randomized controlled trials. PLoS One. 2010;5:e13755. doi:10.1371/journal.pone.0013755. PMID:21060780
  • Traynor K. First quadrivalent flu vaccine approved. Am J Health-Syst Pharm. 2012;69:538.
  • Block SL, Yi T, Sheldon E, Dubovsky F, Falloon J. A randomized, double-blind noninferiority study of quadrivalent live attenuated influenza vaccine in adults. Vaccine. 2011;29:9391–7. doi:10.1016/j.vaccine.2011.09.109. PMID:21983154
  • Nigwekar PV, Kumar A, Padbidri VV, Choudhury A, Chaudhari AB, Kulkarni PS. Safety of Russian-Backbone Trivalent, Live Attenuated Seasonal Influenza Vaccine in Healthy Subjects: Open-Label, Non-randomized Phase 4 Study. Drug safety. 2017.
  • Glück R, Mischler R, Durrer P, Fürer E, Lang AB, Herzog C, et al. Safety and immunogenicity of intranasally administered inactivated trivalent virosome-formulated influenza vaccine containing Eschevichia coli Heat-Labile Toxin as a Mucosal Adjuvant. J Infect Dis. 2000;181:1129–32. doi:10.1086/315337. PMID:10720540
  • Glück U, Gebbers J-O, Glück R. Phase 1 evaluation of intranasal virosomal influenza vaccine with and without Escherichia coli heat-labile toxin in adult volunteers. J Virol. 1999;73:7780–6. PMID:10438868
  • Mutsch M, Zhou W, Rhodes P, Bopp M, Chen RT, Linder T, Spyr C, Steffen R. Use of the inactivated intranasal influenza vaccine and the risk of Bell's palsy in Switzerland. N Eng J Med. 2004;350:896–903. doi:10.1056/NEJMoa030595. PMID:14985487
  • Nicolas JF, Guy B. Intradermal, epidermal and transcutaneous vaccination: from immunology to clinical practice. Exp Rev Vaccines. 2008;7:1201–14. doi:10.1586/14760584.7.8.1201. PMID:18844594
  • Henri S, Guilliams M, Poulin LF, Tamoutounour S, Ardouin L, Dalod M, Malissen B. Disentangling the complexity of the skin dendritic cell network. Immunol Cell Biol. 2010;88:366–75. doi:10.1038/icb.2010.34.
  • Doebel T, Voisin B, Nagao K. Langerhans cells–the macrophage in dendritic cell clothing. Trends Immunol. 2017;38(157):817–28. doi:10.1016/j.it.2017.06.008. PMID:28720426
  • Figdor CG, van Kooyk Y, Adema GJ. C-type lectin receptors on dendritic cells and Langerhans cells. Nat Rev Immunol. 2002;2:77–84. doi:10.1038/nri723. PMID:11910898
  • Belyakov IM, Hammond SA, Ahlers JD, Glenn GM, Berzofsky JA. Transcutaneous immunization induces mucosal CTLs and protective immunity by migration of primed skin dendritic cells. J Clin Invest 2004;113:998–1007. doi:10.1172/JCI20261. PMID:15057306
  • Pulit-Penaloza JA, Esser ES, Vassilieva EV, Lee JW, Taherbhai MT, Pollack BP, Prausnitz MR, Compans RW, Skountzou I. A protective role of murine langerin(+) cells in immune responses to cutaneous vaccination with microneedle patches. Sci Rep. 2014;4:6094. doi:10.1038/srep06094. PMID:25130187
  • Butler D. Measles by the numbers: A race to eradication. Nature. 2015;518:148–9. doi:10.1038/518148a. PMID:25673392
  • Shah V, Choudhury BK. Fabrication, physicochemical characterization, and performance evaluation of biodegradable polymeric microneedle patch system for enhanced transcutaneous flux of high molecular weight therapeutics. AAPS PharmSciTech. 2017;18(8):2936 01502948. doi:10.1208/s12249-017-0774-5. PMID: 28432615
  • Tu J, Du G, Reza Nejadnik M, Monkare J, van der Maaden K, Bomans PHH, Sommerdijk NAJM, Slütter B, Jiskoot W, Bouwstra JA, et al. Mesoporous Silica Nanoparticle-Coated Microneedle arrays for intradermal antigen delivery. Pharm Res. 2017;34(8):1693–706. doi:10.1007/s11095-017-2177-4. PMID:28536970
  • Boks MA, Unger WW, Engels S, Ambrosini M, Kooyk Y, Luttge R. Controlled release of a model vaccine by nanoporous ceramic microneedle arrays. Int J Pharm. 2015;491:375–83. doi:10.1016/j.ijpharm.2015.06.025. PMID:26116016
  • Kim SJ, Shin JH, Noh JY, Song CS, Kim YC. Development of the novel coating formulations for skin vaccination using stainless steel microneedle. Drug Deliv Transl Re. 2016;6:486–97. doi:10.1007/s13346-016-0321-z.
  • Ayittey PN, Walker JS, Rice JJ, de Tombe PP. Glass microneedles for force measurements: a finite-element analysis model. Pflug Arch Eur J Phy. 2009;457:1415–22. doi:10.1007/s00424-008-0605-3.
  • Nguyen HX, Banga AK. Fabrication, characterization and application of sugar microneedles for transdermal drug delivery. Ther Deliv. 2017;8:249–64. doi:10.4155/tde-2016-0096. PMID:28361607
  • Caffarel-Salvador E, Brady AJ, Eltayib E, Meng T, Alonso-Vicente A, Gonzalez-Vazquez P, Torrisi BM, Vicente-Perez EM, Mooney K, Jones DS, et al. Hydrogel-forming microneedle arrays allow detection of drugs and glucose In Vivo: Potential for use in diagnosis and therapeutic drug monitoring. Plos One. 2015;10(10)12:e0145644. doi:10.1371/journal.pone.0145644. PMID: 26717198
  • Hoang MT, Ita KB, Bair DA. Solid microneedles for transdermal delivery of amantadine hydrochloride and pramipexole dihydrochloride. Pharmaceutics. 2015;7:379–96. doi:10.3390/pharmaceutics7040379. PMID:26426039
  • Baek SH, Shin JH, Kim YC. Drug-coated microneedles for rapid and painless local anesthesia. Biomed Microdevices. 2017;19:2. doi:10.1007/s10544-016-0144-1. PMID:28070698
  • Ito Y, Hamasaki N, Higashino H, Murakami Y, Miyamoto N, Takada K. Method to increase the systemically delivered amount of drug from dissolving microneedles. Chem Pharm Bull. 2013;61:8–15. doi:10.1248/cpb.c12-00468.
  • Yu W, Jiang G, Liu D, Li L, Chen H, Liu Y, Huang Q, Tong Z, Yao J, Kong X. Fabrication of biodegradable composite microneedles based on calcium sulfate and gelatin for transdermal delivery of insulin. Mater Sci Eng C Mater Biol Appl. 2017;71:725–34. doi:10.1016/j.msec.2016.10.063.
  • Wiggan O, Livengood JA, Silengo SJ, Kinney RM, Osorio JE, Huang CYH, Stinchcomb DT. Novel formulations enhance the thermal stability of live-attenuated flavivirus vaccines. Vaccine. 2011;29:7456–62. doi:10.1016/j.vaccine.2011.07.054. PMID:21803103
  • Gay NJ. The theory of measles elimination: implications for the design of elimination strategies. J Infect Dis. 2004;189 Suppl 1:S27–35. doi:10.1086/381592. PMID:15106086
  • Edens C, Collins ML, Ayers J, Rota PA, Prausnitz MR. Measles vaccination using a microneedle patch. Vaccine. 2013;31:3403–9. doi:10.1016/j.vaccine.2012.09.062. PMID:23044406
  • Li N, Wang N, Wang XT, Zhen YY, Wang T. Microneedle arrays delivery of the conventional vaccines based on nonvirulent viruses. Drug Delivery. 2016;23:3234–47. doi:10.3109/10717544.2016.1165311. PMID:26967666
  • Edens C, Collins ML, Goodson JL, Rota PA, Prausnitz MR. A microneedle patch containing measles vaccine is immunogenic in non-human primates. Vaccine 2015;33:4712–8. doi:10.1016/j.vaccine.2015.02.074. PMID:25770786
  • Ding Z, Verbaan FJ, Bivas-Benita M, Bungener L, Huckriede A, van den Berg DJ, Kersten G, Bouwstra JA. Microneedle arrays for the transcutaneous immunization of diphtheria and influenza in BALB/c mice. J Controlled Release. 2009;136:71–8. doi:10.1016/j.jconrel.2009.01.025. PMID:19331846
  • Vrdoljak A, McGrath MG, Carey JB, Draper SJ, Hill AVS, O'Mahony C, Crean AM, Moore AC. Coated microneedle arrays for transcutaneous delivery of live virus vaccines. J Controlled Release. 2012;159:34–42. doi:10.1016/j.jconrel.2011.12.026. PMID:22245683
  • Kim E, Erdos G, Huang SH, Kenniston T, Falo LD, Gambotto A. Preventative vaccines for Zika Virus outbreak: Preliminary evaluation. Ebiomedicine. 2016;13:315–20. doi:10.1016/j.ebiom.2016.09.028. PMID:27717627
  • Carey JB, Vrdoljak A, O'Mahony C, Hill AVS, Draper SJ, Moore AC. Microneedle-mediated immunization of an adenovirus-based malaria vaccine enhances antigen-specific antibody immunity and reduces anti-vector responses compared to the intradermal route. Sci Reports. 2014;4;6154. doi:10.1038/srep06154. PMID:25142082
  • Wang YH, Vlasova A, Velasquez DE, Saif LJ, Kandasamy S, Kochba E, Levin Y, Jiang B. Skin Vaccination against Rotavirus Using Microneedles: Proof of Concept in Gnotobiotic Piglets. Plos One. 2016;11:e0166038. doi:10.1371/journal.pone.0166038. PMID:27824918
  • Muller DA, Pearson FE, Fernando GJP, Agyei-Yeboah C, Owens NS, Corrie SR, Crichton ML, Wei JC, Weldon WC, Oberste MS, et al. Inactivated poliovirus type 2 vaccine delivered to rat skin via high density microprojection array elicits potent neutralising antibody responses. Sci Reports. 2016;6:22094. doi:10.1038/srep22094. PMID:26911254
  • Weldon WC, Martin MP, Zarnitsyn V, Wang BZ, Koutsonanos D, Skountzou I, Prausnitz MR, Compans RW. Microneedle vaccination with stabilized recombinant influenza virus hemagglutinin induces improved protective immunity. Clin Vaccine Immunol. 2011;18:647–54. doi:10.1128/CVI.00435-10.
  • Kommareddy S, Baudner BC, Bonificio A, Gallorini S, Palladino G, Determan AS, Dohmeier DM, Kroells KD, Sternjohn JR, Singh M, et al. Influenza subunit vaccine coated microneedle patches elicit comparable immune responses to intramuscular injection in guinea pigs. Vaccine 2013;31:3435–41. doi:10.1016/j.vaccine.2013.01.050. PMID:23398932
  • Mikszta JA, Dekker JP, Harvey NG, Dean CH, Brittingham JM, Huang J, Sullivan VJ, Dyas B, Roy CJ, Ulrich RG. Microneedle-based intradermal delivery of the anthrax recombinant protective antigen vaccine. Infect Immunity. 2006;74:6806–10. doi:10.1128/IAI.01210-06. PMID:17030580
  • Song JM, Kim YC, Lipatov AS, Pearton M, Davis CT, Yoo DG, Park KM, Chen LM, Quan FS, Birchall JC, et al. Microneedle delivery of H5N1 influenza virus-like particles to the skin induces Long-Lasting B- and T-Cell responses in mice. Clin Vaccine Immunol. 2010;17:1381–9. doi:10.1128/CVI.00100-10. PMID:20631330
  • Kim MC, Lee JW, Choi HJ, Lee YN, Hwang HS, Lee J, Kim C, Lee JS, Montemagno C, Prausnitz MR, et al. Microneedle patch delivery to the skin of virus-like particles containing heterologous M2e extracellular domains of influenza virus induces broad heterosubtypic cross-protection. J Controlled Release. 2015;210:208–16. doi:10.1016/j.jconrel.2015.05.278. PMID:26003039
  • Hooper JW, Golden JW, Ferro AM, King AD. Smallpox DNA vaccine delivered by novel skin electroporation device protects mice against intranasal poxvirus challenge. Vaccine. 2007;25:1814–23. doi:10.1016/j.vaccine.2006.11.017. PMID:17240007
  • Alarcon JB, Hartley AW, Harvey NG, Mikszta JA. Preclinical evaluation of microneedle technology for intradermal delivery of influenza vaccines. Clin Vaccine Immunol. 2007;14:375–81. doi:10.1128/CVI.00387-06. PMID:17329444
  • Song JM, Kim YC, O EJ, Compans RW, Prausnitz MR, Kang SM. DNA vaccination in the skin using microneedles improves protection against influenza. Mol Therapy. 2012;20:1472–80. doi:10.1038/mt.2012.69. PMID:22508490
  • Matsuo K, Hirobe S, Yokota Y, Ayabe Y, Seto M, Quan YS, Kamiyama F, Tougan T, Horii T, Mukai Y, et al. Transcutaneous immunization using a dissolving microneedle array protects against tetanus, diphtheria, malaria, and influenza. J Controlled Release. 2012;160:495–501. doi:10.1016/j.jconrel.2012.04.001. PMID:22516091
  • Perdue ML, Arnold F, Li S, Donabedian A, Cioce V, Warf T, Huebner R. The future of cell culture-based influenza vaccine production. Exp Rev Vaccin. 2011;10:1183–94. doi:10.1586/erv.11.82. PMID:21854311
  • Endmann A, Kluender K, Kapp K, Riede O, Oswald D, Talman EG, Schroff M, Kleuss C, Ruiters MH, Juhls C. Cationic lipid-formulated DNA vaccine against Hepatitis B virus: Immunogenicity of MIDGE-Th1 vectors encoding small and large surface antigen in comparison to a licensed protein vaccine. Plos One. 2014;9(7):e101715. doi:10.1371/journal.pone.0101715. PMID:24992038
  • Cole G, McCaffrey J, Ali AA, McBride JW, McCrudden CM, Vincente-Perez EM, et al. Dissolving microneedles for DNA vaccination: Improving functionality via polymer characterization and RALA complexation. Hum Vaccin Immunother. 2017;13:50–62. doi:10.1080/21645515.2016.1248008.
  • Qiu YQ, Guo L, Zhang SH. DNA-based vaccination against hepatitis B virus using dissolving microneedle arrays adjuvanted by cationic liposomes and CpG ODN (vol 23, pg 2391, 2015). Drug Delivery. 2016;23:3179. PMID:26679752
  • Kim NW, Lee MS, Kim KR, Lee JE, Lee K, Park JS, Matsumoto Y, Jo DG, Lee H, Lee DS, et al. Polyplex-releasing microneedles for enhanced cutaneous delivery of DNA vaccine. J Controlled Release. 2014;179:11–7. doi:10.1016/j.jconrel.2014.01.016. PMID:24462900
  • Yang HW, Ye L, Guo XD, Yang CL, Compans RW, Prausnitz MR. Ebola vaccination using a DNA vaccine coated on PLGA-PLL/gamma PGA nanoparticles administered using a microneedle patch. Adv Healthc Mater. 2017;6(1):1600750. doi:10.1002/adhm.201600750.
  • Seok H, Noh JY, Lee DY, Kim SJ, Song CS, Kim YC. Effective humoral immune response from a H1N1 DNA vaccine delivered to the skin by microneedles coated with PLGA-based cationic nanoparticles. J Control Release. 2017;265:66–74. doi:10.1016/j.jconrel.2017.04.027. PMID:28434892
  • Arya JM, Dewitt K, Scott-Garrard M, Chiang YW, Prausnitz MR. Rabies vaccination in dogs using a dissolving microneedle patch. J Controlled Release. 2016;239:19–26. doi:10.1016/j.jconrel.2016.08.012. PMID:27524283
  • Kim YC, Yoo DG, Compans RW, Kang SM, Prausnitz MR. Cross-protection by co-immunization with influenza hemagglutinin DNA and inactivated virus vaccine using coated microneedles. J Controlled Release 2013;172:579–88. doi:10.1016/j.jconrel.2013.04.016. PMID:23643528
  • Rouphael NG, Paine M, Mosley R, Henry S, McAllister DV, Kalluri H, Pewin W, Frew PM, Yu T, Thornburg NJ, et al. The safety, immunogenicity, and acceptability of inactivated influenza vaccine delivered by microneedle patch (TIV-MNP 2015): a randomised, partly blinded, placebo-controlled, phase 1 trial. Lancet 2017;390:649–58. doi:10.1016/S0140-6736(17)30575-5. PMID:28666680
  • Estivariz CF, Snider CJ, Anand A, Hampton LM, Bari TI, Billah MM, Chai SJ, Wassilak SG, Heffelfinger JD, Zaman K. Lessons learned from the introduction of inactivated Poliovirus vaccine in Bangladesh. J Infect Dis. 2017;216:S122–S9. doi:10.1093/infdis/jiw510. PMID:28838154
  • Mudie DM, Amidon GL, Amidon GE. Physiological parameters for oral delivery and in vitro testing. Mol Pharm 2010;7:1388–405. doi:10.1021/mp100149j. PMID:20822152
  • Posgai AL, Wasserfall CH, Kwon KC, Daniell H, Schatz DA, Atkinson MA. Plant-based vaccines for oral delivery of type 1 diabetes-related autoantigens: Evaluating oral tolerance mechanisms and disease prevention in NOD mice. Sci Rep. 2017;7:42372. doi:10.1038/srep42372. PMID:28205558
  • Price DN, Kusewitt DF, Lino CA, McBride AA, Muttil P. Oral tolerance to environmental Mycobacteria interferes with Intradermal, but not pulmonary, immunization against Tuberculosis. PLoS Pathog. 2016;12:e1005614. doi:10.1371/journal.ppat.1005614. PMID:27153120
  • Wang T, Zhen Y, Ma X, Wei B, Li S, Wang N. Mannosylated and lipid A-incorporating cationic liposomes constituting microneedle arrays as an effective oral mucosal HBV vaccine applicable in the controlled temperature chain. Colloids Surf B Biointerfaces. 2015;126:520–30. doi:10.1016/j.colsurfb.2015.01.005. PMID:25612819