Sunteți pe pagina 1din 20

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/289502724

Nanoparticulate mediated transcutaneous immunization: Myth or reality

Article  in  Nanomedicine: nanotechnology, biology, and medicine · January 2016


DOI: 10.1016/j.nano.2015.12.372

CITATIONS READS

9 187

6 authors, including:

Monika Kaurav Sunita Minz


Guru Ghasidas University Indira Gandhi National Tribal University
6 PUBLICATIONS   19 CITATIONS    9 PUBLICATIONS   48 CITATIONS   

SEE PROFILE SEE PROFILE

Kantrol Sahu Manoj Kumar


Guru Ghasidas University NBA SCHOOL OF BUSINESS NEW DELHI
7 PUBLICATIONS   24 CITATIONS    113 PUBLICATIONS   2,144 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

DESIGN AND OPTIMIZATION OF NANO VACCINES, VACCINE DELIVERY THROUGH NASAL AND PULMONARY ROUTES View project

Nanoparticles View project

All content following this page was uploaded by Monika Kaurav on 19 July 2016.

The user has requested enhancement of the downloaded file.


Nanomedicine: Nanotechnology, Biology, and Medicine
12 (2016) 1063 – 1081

Original Article nanomedjournal.com

Nanoparticulate mediated transcutaneous immunization: Myth or reality


Monika Kaurav, MPharm a , Sunita Minz, MPharm a , Kantrol Sahu, MPharm a , Manoj Kumar, PhD a ,
Jitender Madan, PhD b , Ravi Shankar Pandey, PhD a,⁎
a
SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
b
Chandigarh College of Pharmacy, Mohali, Panjab, India
Received 2 October 2015; accepted 17 December 2015

Abstract

Transcutaneous immunization (TCI) is a promising route of vaccine delivery through skin due to many well documented advantages. The
main obstacle in TCI is the skin's top dead layer i.e. stratum corneum which is difficult to penetrate. Efficiently delivery of antigen to the
immune competent cells of epidermis or dermis in TCI might elicit an effective immune response. In this review, skin immunology with a
particular focus on potential of immunological active receptors in influencing adaptive immune responses is highlighted. The challenges with
TCI and methods to improve it using different adjuvants, chemical and physical approaches, delivery systems, and combination of above
methods to further improve immune response following skin application of antigen are elaborately discussed. Nanoparticulate vaccine
delivery systems with reference to their applications in TCI are classified according to their chronological development. Conclusively,
clinical translations of above methods are also briefly reviewed.

From the Clinical Editor: Transcutaneous immunization has been investigated by many as a promising route of vaccination. In this
comprehensive review article, the authors described and discussed the existing knowledge and difficulties in this approach. Furthermore,
ways of improving transcutaneous delivery were also reviewed.
© 2016 Elsevier Inc. All rights reserved.

Key words: Transcutaneous immunization; Adjuvants; Nanoparticulate delivery systems

Vaccination has been one of the extensively booming medical Traditionally available vaccine delivery methods are oral
interventions in diminution of infectious diseases since the (OR), intranasal (IN), intradermal (ID), intramuscular (IM) and
nineteenth century. 1 According to definition, a good vaccine subcutaneous (SC). 4 Out of these IM, SC and ID immunizations
should be safe, causes minimal pain if invasive and most are classified as needle-based techniques. Although needle-based
importantly elicits long lasting strong and protective immune techniques are effective in achieving the desired immune
response. 2 Activation of immune system is the main concern to response, however their use has numerous limitations such as
be faced in vaccine development. Due to the limited accessibility injection requires sterilized needles and skilled people. More-
of efficiently active antigen presenting cells (APCs) at the over, injection can be irritating and painful, particularly in
location of vaccination of traditional vaccine method, represen- children. 5 The muscular site, a conventional site of immuniza-
tation of vaccine originated antigen to the immune system is tion, is not regarded as to be a proficient site for antigen
reduced which makes it insufficient process. 3 presentation owing to lack of suitable density of APCs dendritic
cells (DCs) or macrophages. 6 However, antigens alone usually
are inadequately immunogenic and hence necessitate to be
prepared with adjuvants to amplify DC establishment and
infiltration into the intramuscular immunization sites, 6 -9 finally
Conflicts of interests: The authors declare no conflict of interest. from all above, adding to the belief that the muscles and SC
⁎Corresponding author at: SLT Institute of Pharmaceutical Sciences,
routes are not an ideal site meant for immunization. The
Guru Ghasidas Vishwavidyalaya, Bilaspur, India. drawbacks of injectable vaccines have heighten the research on
E-mail addresses: monika11kaurav@gmail.com (M. Kaurav),
sunitaminz.3481@rediffmail.com (S. Minz), kantrol.sahu23@gmail.com
oral, 10 nasal, 11 pulmonary 12 and transcutaneous 13 deliveries of
(K. Sahu), mrmanojkumar1@yahoo.co.in (M. Kumar), vaccines. Despite the efficacy of traditional (attenuated or killed)
jitenderpharmacy@gmail.com (J. Madan), vaccines, there is concern over their safety, 14 represented in
ravishankarpandey2@rediffmail.com, rspandey1@gmail.com (R.S. Pandey). Figure 1, along with limitation of injectable vaccines. 15
http://dx.doi.org/10.1016/j.nano.2015.12.372
1549-9634/© 2016 Elsevier Inc. All rights reserved.

Please cite this article as: Kaurav M, et al, Nanoparticulate mediated transcutaneous immunization: Myth or reality. Nanomedicine: NBM 2016;12:1063-1081,
http://dx.doi.org/10.1016/j.nano.2015.12.372
1064 M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081

Figure 1. Disadvantages of traditional vaccines and limitations of injectable vaccines.

Skin is abundant in trained APCs such as DCs in the dermis stratum corneum, outermost layer of the skin, and indicate a
and Langerhans cells (LCs) in the epidermis, these cells can system of immune cells that bring about 25% of the total skin
professionally activate both in vitro and in vivo primary immune area and can transport antigen from the skin into the lymph node
responses. 16,17 Thus TCI is one of the most promising to elicit immune responses. 27 The immune cells and immune
immunization methods. Vaccine administration via the skin responses of skin immune system are represented in Figure 2.
without any pain by means of a larger surface area is referred to
as transcutaneous immunization (TCI). The transcutaneous route
is principally striking because the skin is vastly accessible and DCs as a link between innate and adaptive immunity
has distinctive immunological characteristics, 18 while for a long
time, it has been acknowledged that an effectual immune As discussed before, skin DCs are an important linkage
response can be induced through the skin and various diverse between innate and adaptive immunity. They tutor and excite B
approaches have been attempted. One of victorious examples of and T lymphocytes with the contribution of central role in both,
TCI is a scarifying skin in the case of vaccinia virus vaccination cell mediated as well as humoral immunity. Adaptive immunity
in humans. 19 The existence of skilled APCs in the epidermis and starts when DC cells matured and migrated to lymph node where
the dermis mediates the immune response following cutaneous it interacts with T cell, resulted in the proliferation of B and T
immunization. 16 An additional crucial rationale for considering lymphocytes in the spleen and lymph nodes that are secondary
the transcutaneous route is the prospective for protected immune lymphoid organs. Both these cells are developed from common
stimulation, as it circumvents the straight contact between lymphocyte progenitor in bone marrow. DCs are another major
persuasive adjuvants with the general circulation. 20 The imperative APC in the skin and take part in a vital function in
advantages associated with TCI listed in Table 1. 21,22 both the innate and adaptive immune responses. Immature DCs
are activated by agents such as microbes, cells of immune
systems. Pathogen recognition receptors (PRRs) are agents that
Immunological mechanism system of skin and initiation of initiated these responses by binding to them. Even though PRRs
immune response are expressed on several cell types, DCs have an important role
in controlling immune response, so focus of DC activation
Foremost components of skin immune system are keratino- concentrated on PRRs. 17 The mechanism of action of PRRs
cytes in the viable epidermis while melanocytes, Merkel cells upon binding of an antigen with adjuvant is shown in Figure 3.
and Langerhans cells are less abundant and present in dermis. PRRs are further divided into three major classes, RIG-I-like
Keratinocytes constitute about 90% of total epidermal cell receptors (RLRs), NOD-like receptors (NLRs) and Toll-like
population playing important role in innate immunity of the skin. receptors (TLRs). 28
In case of skin barrier interruption, keratinocytes produce many TLRs are one of the mainly imperative PRRs and provide
types of chemokines, cytokines and antimicrobial proteins and defense to host against a range of pathogens. On the basis of
peptides [eg. tumor necrosis factor-α (TNF-α), interleukin-1α selective recognition to diverse bacterial, viral pathogen or
(IL-1α), interleukin-1β (IL-1β) and granulocyte macrophage- endogenous signals, ten TLR members have been identified in
colony stimulating factor (GM-CSF)]. 23 These chemokines are human. 29 These TLRs on the basis of their ligand recognition
key mediators in the generation of innate immunity upon and localization either on cell surface or intracellular, can be
interaction with DCs. 24 Besides keratinocytes, other cells further divided into subfamilies. In this subfamily of TLR 3, 7, 8
involved in cytokine secretion and DC maturation are natural and 9 recognize nucleic acids, whereas the subfamilies of TLRs
killer cells, mast cells, macrophages, and neutrophils. 25 The skin 1, 2, 4 and 6 recognize lipids. 30 TLRs 1, 2, 4, 5, 6, 10 and 11 that
in addition has significant immunological roles owing to the detect bacterial products other than nucleic acid are present on
presence of the distinctive skin-associated lymphoid tissues the cell surface, whereas TLRs 3, 7, 8 and 9 that detect nucleic
(SALT), 26 which is constituted by skilled APCs, such as LCs acids are located intracellularly, either on late endosomes or
and dDCs (dermal dendritic cells) along with other skin cell lysosomes. Such limited localization possibly will afford the
subsets of T-lymphocytes and lymph nodes, although of the total mechanism by which DCs evade unprompted activation by self
skin cells, LCs detail for only 1% of those present near the nucleic acids. 31
M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081 1065

Table 1
Advantages of transcutaneous vaccines. 21,22
Non-invasive Does not require use of needle
Avoid rise of infection Avoid problem of blood transmissible infection due to needle re-use associated with injectable vaccine
Stable No first pass metabolism thus provide protection to antigen from enzymatic digestion
Better patient compliance Self administration possible, no need of trained personnel
Induction of immune response Stimulation of innate immune system that trigged appropriate adaptive immune response and induction of immunological
memory, activates LCs and DCs which induce Th1, Th2, cytotoxic T lymphocytes and different subclasses of IgG, IgA etc
No strict purity concern Given via skin route not in systemic so sterilization of formulation is not required
Easy and fast to produce Mass production of skin vaccine may be less expensive than that of injectable vaccine that requires high standard of purity
with sterility
Low cost of production Low cost due to no strict purity and sterility, increase usage and demand in developing countries
Protective barrier function Avoid risk of invasion by microorganisms, prevention of pathogen entry Into systemic circulation by local immune response
and prevention of infection spread
Effective in children, in aged and Easy withdrawal possible in case of side/adverse effect as compared to injectable vaccine and more attractive due to self
immunocompromised persons applicable

Most studies which are related to skin focused on TLR size which limit their transport across stratum corneum. 40 Apart
expression on LCs and dDCs, which is divergent and from this, a number of adjuvants are too toxic in nature for
furthermore diverges from further subtypes of DCs at mucosal clinical use. Adjuvants established with TCI are bacterial
outside or in the blood circulation. The TLRs and their natural exotoxins which are responsible for different cytokines expres-
ligand location on skin immune cells (human and mouse) are sion from re-stimulated lymphocytes and TLR ligands. 41,42 To
listed in Table 2. 29,31-34 The TLR distribution on DCs and the elicit strong immune response with reduced nonspecific adverse
difference in the epithelia micromilieu may persuade the immune inflammatory response, there must be a need of optimal adjuvant
alteration function of definite adjuvants and in that way the which targets LCs in skin eliciting systemic antibodies including
choice of adjuvants for TCI. 35 IgG and secretory IgA responses in both mice and humans.
Table 3 briefly discusses promising adjuvants established safe in
preclinical trials of TCI. 43 -53
Transcutaneous immunization (TCI)

As revealed before, TCI vaccine delivery through which Skin permeation enhancement approaches for TCI
antigen topically applied on epidermis, now becomes a rising
antigen delivery methodology due to the fact that skin is Protein, peptides and vaccines are not suitable candidates for
exceedingly accessible and has inimitable immunological oral delivery because they are prone to high degree of first pass
distinctiveness. 18 However, topical antigen application perme- metabolism in the liver. Skin as a largest and most accessible
ation through skin into epidermis is restricted by the impervious organ of the body provides a suitable site for administration of
obstruction role of the stratum corneum, the outermost layer of above candidates. Transcutaneous delivery routes offer promis-
epidermis. 36 To induce efficient TCI, main challenges are ing site by target delivery to immune cells such as LCs and
co-administration of adjuvants with antigen(s) and permeation dendritic cells in the skin, can improve immune response and has
through skin barriers to induce strong immune response via fascinating rising significance. Recently, advancements in
uptake of antigen by stimulated DCs, their maturation and pharmaceutical biotechnologies and emerging improved tech-
relocation in an appropriate manner. niques and instruments can overcome problems related with the
stratum corneum barrier. In the direction of overcoming skin
barrier to non-invasive delivery, various sophisticated delivery
Adjuvants used in TCI systems, passive as well as active approaches to optimize
transcutaneous delivery of protein and peptides have been
In immunology “adjuvants” are agents that stimulate the developed. All the vaccine candidates used for TCI administra-
immune system and increase the response to the vaccine, tion do not have ideal physiochemical properties, so there is a
however with or without any antigenic effect in itself. In need to add some chemicals or vehicle or devices or formulations
combination with antigen adjuvant can enhance, accelerate or to help them penetrate or cross the stratum corneum to get the
prolong antigen specific immune response. 37 Adjuvants are one desired therapeutic response. Various approaches are discussed
of most important components in TCI, because response is below (Figure 4):
adjuvant dependent. Co-application of adjuvant with antigen is
required to elicit thriving immune responses by TCI. 38 The
variety of prospective adjuvants for vaccine is wide which Physical approaches
include microbial bacterial and toxin products, oligodeoxynu-
cleotides, cytokines, liposomes and mineral salts. 39 Aluminium As previously reviewed, penetration of substances in the skin
hydroxide, calcium phosphate, stearyl tyrosine and MF 59 is restricted by the SC which acts as a physical barrier, so the
emulsions are undesirable for TCI; due to their relatively large topical application of the vaccine formulation alone restricts its
1066 M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081

Figure 2. Skin immune system. The skin is enriched with immunocompetent cells such as LCs, keratinocytes and several dDCs. Keratinocytes induce innate
immunity. LCs and DCs capture external Ag. Migrate into lymph node where they present Ag to T cells and activate Ag specific T cells and B Cells. Activated B
cells and T cells migrate to other parts and induce Ag-specific immune responses.

passage across the skin and hence efficient immune response is against CT (cholera toxin) were improved when the area of
not achieved. Various physical approaches such as electropora- immunization was swabbed with isopropanol prior to application
tion, iontophoresis, abrasions, tape stripping, sonophoresis, of the antigen. 62 Karande 63 formulated several formulations of
micro needles etc have been investigated to increase the commonly used chemicals, screened their potency (delivery of
penetration of substances across the skin by disrupting SC. antigen) in vitro and subsequently tested for their adjuvancy
Another approach to bypass the barrier function of SC is (activation of immune response) followed by investigation in
application of microneedles. 54 A microneedle array contains vivo for their aptitude to produce antibodies in opposition to
many micrometer-sized needles that can create a transport ovalbumin which is applied topically and shows to be
pathway large enough for proteins and NPs, but small enough to extensively more effectual in producing anti-IgG titers of
avoid harm. 55,56 Tape-stripping method has been shown to ovalbumin. Many delivery systems such as liposomes, dendri-
enhance cytotoxic T cells and cytokine mediated immune mers, and microemulsions because of their supramolecular
responses upon subsequent application of various antigens and structure have also been used as chemical enhancers that increase
adjuvants to the skin in mice. 57 Similarly electrode preparation skin permeability and solubilization of medicaments in the
pads, emery paper, rubbing gauze or porous rock on the skin take formulation and their partitioning into the skin. 64,65 These results
out cells by their scratchy effects and have been found to enhance have shown that formulations prepared with chemical enhancers
immune responses in humans. 58 Mild hyperthermia with can be advantageously used to deliver antigens through the
abrasion enhances transport of antigen into the skin, presents it transcutaneous route.
to LCs, enhances migration of activated LCs to lymph nodes, and
triggers cascade of immune system.
Novel approaches for TCI using particulate antigen delivery
systems
Chemical enhancer approaches
Formulation development of antigens in particulate carrier
Chemical moiety which reduces the barrier function of the systems is a fascinating approach to advance vaccine delivery,
stratum corneum is known as penetration enhancers. The also through the transcutaneous method. 66,67 It is known that
enhancer can either disrupt lipid organization and enhance disruption of skin barrier increases the transcutaneous perme-
drug diffusion coefficient or interact with keratin in corneocytes, ation of antigen and makes it easy to be taken up by antigen
thereby opening the dense protein structure. 59 In addition, via presenting cells. So that non-invasive techniques that are capable
increases of the partitioning of drugs, chemical enhancers result of disrupting the epidermal skin barriers have been widely
in an increased diffusion rate. 60,61 More than 300 penetration investigated to activate the immune system and by cytokines
enhancers are known and categorized into different categories persuade the secretion of pro-inflammatory keratinocytes and
based on their mechanism of penetration enhancement. Pre promote proceeding of immune stimulatory events. Recently a
hydration using alcohol swab can enhance the immune response lot of attention has been focused on micro/nanometric delivery
elicited by TCI. Scharton-Kersten reported that antibody titers systems such as patches, nanoparticles (NPs), liposomes,
M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081 1067

Figure 3. Mechanism of action of pattern recognition receptors (PRR) upon binding of antigen with adjuvants used in TCI.

transferosomes, solution, emulsions, suspensions, niosomes, inner aqueous core. 68 Liposomes' main component, phospho-
cubosomes etc. Main priority is to formulate formulation to lipids are biocompatible and eco-friendly and these qualities
target antigen to APCs and to protect antigen from degradation make liposomes a protected system, competent to be used in the
for prolonged and long lasting effect. pharmaceutical ground. 69 They can result in prolonged primary
In present review, advanced and novel topical delivery activation of T cells in vivo via averting the degradation of
systems have been categorized into three generations of antigens. 67 To deliver the molecule at the site of action, the lipid
chronological development depending upon their utilization for bilayers fuse with other bilayer such as cell membrane delivering
topical vaccination purpose. First generation topical delivery the liposomal contents. 70 Tyagi et al, 71 reported the immu-
systems have seen a sturdy increase in clinical use for delivery of ne-enhancing effects of elastic liposomes consisting merozoite
vaccines that are permissible to the skin via passive diffusion. surface protein-1 of Plasmodium falciparum through transcuta-
Work in this area progressed before 90s such as liposomes, neous delivery. Mishra et al, 72 have studied that hepatitis B
polymerosomes, niosomes NPs, patches etc. Second generation surface antigen (HBsAg)-loaded elastic liposomes were an
delivery systems include systems with some more advancement effective mode for enhanced immunity against the antigen via
which emerged in 1990 to 2005 such as transferosomes, transcutaneous route.
ethosomes, cubosomes, vesosomes, immunostimulatory com-
plexes (ISCOMs), dendrimers etc. Sometimes they are used
along with chemical enhancers to enhance permeability and Nanoparticles (NPs)
drive transcutaneous transport and have also led to clinical NPs are striking means for transcutaneous antigen delivery.
products. Third generation delivery systems emerge from 2005 The underlying mechanism is that by disrupting SC as a result,
to present and include inorganic NPs such as gold, magnetic, nano-bio interaction with skin lipid, the antigen encapsulated in
quantum dots, carbon nanotubes etc. Schematic diagram of NPs delivered to the SC into the skin via induction of transient
several transcutaneous antigen delivery technologies for TCI is and reversible opening of the stratum corneum. Just because they
shown in Figure 5 and briefly listed about different generation seem alike in structure and size to the natural pathogen, which
delivery systems consisting about mechanism of action, are dynamically taken up by the APCs, can able to improve
properties, limitation along with their utilization for TCI in antigen uptake. 73 An additional lead is the prospect to
Table 4. encapsulate antigen and adjuvant together in the same particle,
which is put forward to improve the immunogenicity. 74
Additionally, it was proved that NP vaccine can penetrate hair
First generation delivery systems (before 90s) follicles where high density of APCs are present and target the
Liposomes carried antigen toward APCs ultimately stimulate immune
responses. 75,76 Several numbers of polymers have been reported
Liposomes are spherical artificial phospholipid vesicles. They for preparing NPs and induce well-built TCI. Schematic diagram
assemble impetuously into bilayered arrangements including an shows generation of adaptive immune response following topical
1068 M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081

Table 2 interacting with a negative charge in the skin to improve antigen


Toll like receptors with their natural ligands and localities. 29,31-34 diffusion into the deeper layer of skin. 82 Chitosan and its
TLR Ligands Ligand locality derivative have several advantageous properties such as
TLR1 + TLR2 Tri-acyl lipopeptides Bacteria
biodegradability, biocompatibility, low toxicity and mild
TLR2 + TLR6 Di-acyl lipopeptides Mycoplasm preparation method made chitosan ideal for development of
TLR2 Glycolipids Bacteria drug delivery system. 83,84 N-trimethyl chitosan (TMC) deriva-
Lipoteichoic acid Bacteria tive of chitosan is water soluble and bears a permanent positive
HSP60, 70 and 90, grp96 Host cells a charge over a wide pH range which prominently increases the
TLR3 Double-stranded virus antigen transdermal penetration in vitro and in vivo. 85 TMC
RNA (dsRNA)
could appreciably modify the secondary structure arrangement of
TLR4 LPS Gram negative
bacteria keratin in the stratum corneum, increase the water content in the
HSPs Bacteria and host cells stratum corneum, enhance cell membrane fluidity, has intrinsic
Fibrinogen, heparin Host cells a adjuvant properties and trigger DC maturation. 86 Several reports
sulfate fragment demonstrated that chitosan NPs act as adjuvant, can also function
Hyaluronic acid as a depot 87 and are efficiently taken up by DCs than plain
fragment, β-defensin antigen and lead to the higher immune response than
TLR5 Flagellin Bacteria
TLR7/8 Single stranded RNA (ssRNA) Virus
intramuscular injection. 18 Li et al, 88 prepared chitosan NPs
TLR9 Unmethylated CpG DNA Bacteria loaded with ovalbumin or gp 100 protein which has shown
TLR10 Unidentified Unidentified significantly higher anti-OVA IgG titers and stimulated tumor
TLR11 (mice only) Profilin Toxoplasma gondii immune response for respective NPs after TCI.
TLR12 (mice only) Unidentified Unidentified
TLR13 (mice only) Unidentified Unidentified Niosomes
a
Danger associated signals molecules (DAMPs).
Niosomes are another type of delivery system that has been
reported in application of noninvasive TCI, having similar
physical properties and preparation method that of
application of antigen containing NPs with or without adjuvant liposomes. 89,90 For target delivery to the LCs niosomes made
(Figure 6). up of Span 60, Span 85, cholesterol and stearylamine containing
BSA were coated with O-palmitoyl mannan which is a modified
Polymeric PLA/PLGA NPs
polysaccharide. This niosomal formulation has shown notable
Commonly used PLGA/PLA NPs are extensively studied by higher level of serum IgG titers in compared to alum absorbed
several investigators for transcutaneous antigen delivery. When BSA and plain uncoated niosomes upon TCI administration, but
applied to the skin nano- and micron sized particulate carriers are less than those achieved with an equivalent dose of BSA-alum
mount up in the follicle ends, sebaceous glands or skin layers. after IM injection. 91 Maheshwari et al, 92 reported that cholera
The recent work of Hansen and Lahr 77 has shown that massaging toxin B when co-administered with the niosomes loaded with
of aqueous NP suspension as well as a hydrogel formulation of HBsAg topically is a potential adjuvant for cutaneous immuni-
NPs, into pig ear skin in vitro permeates in deep layers and into zation and it also explored niosomes vaccine delivery systems
hair follicles more compared to simple aqueous solution. They can be effectual as topical delivery of vaccines. Van den Bergh et
prepared NPs with polylactic-co-glycolic acid (PLGA) which is al, 93 prepared many formulations of surfactant-based elastic
biocompatible and biodegradable polymer along with a stabilizer vesicles, made up of a sucrose-laurate ester which is a
polyvinyl alcohol. Furthermore such particles may efficiently bilayer-forming surfactant and octaoxyethylene-laurate ester
able to release diverse loaded compounds that enter in the also a surfactant and sodium bis-tri-decyl sulfo-succinate
skin. 78,79 Mattheolabakis et al, 80 examined the potential of OVA which is a charge inducer. Ding et al, 94 prepared pH sensitive
loaded PLA NPs as a vector for antigen delivery via diphtheria toxoid loaded elastic vesicles. Results have shown
transcutaneous route to induce efficient cytokine production. In that vesicles that had antigen preserving potential and release
vitro culture of splenocytes with OVA re-stimulate thus elicited a antigen at desire pH make them excellent carrier for future
little higher levels of IFN-γ and IL-2 (P b 0.001) in mice, studies.
compared to those immunized with OVA in solution. Vogt et
al, 76 further investigated that smaller size NPs (40 nm NPs, but
Patches
not 750 or 1500 nm NPs) may be efficiently used for
transcutaneous delivery of vaccine compounds via the hair
Needle-free delivery has become a global priority because of
follicle into cutaneous APCs.
the hazard of needle-borne diseases associated with re-use and
Chitosan NPs inappropriate disposal of needle. Patch based vaccination has
been demonstrated for both promoting the penetration of
The properties of chitosan and its derivatives are promising in antigenic proteins across the stratum corneum and inducing
the application of TCI. Recently, they are reported as potential Ag-specific humoral immune responses, which neutralizes viral
transdermal penetration enhancer and their NPs were reported as infection and bacterial toxins in a safe manner. The underlying
an immune enhancer in TCI. 81 Chitosan appeared to be mechanism is hypothetical in that the antigenic proteins present
Table 3
Immune adjuvants used in transcutaneous immunization.
TCI adjuvants/resources Molecular mechanism Functions in TCI Benefits Safety/immunogenicity Reference
43
CT, CT-B A non hazardous B subunit with Raise LCs population in epidermis; Augmentation of level of Typical mucosal adjuvants, competent
strong binding affinity on GM 1 induce antibody responses and IgA in serum and mucosa in several animal models, but toxic in
ganglioside receptors, which Th 1 and Th 2 mediated cytokines humans
presents on skin and mucosal production; CT-B potentiate
epithelial cells, lymphocytes and DCs Th1 related immune responses;
produce ILs 2,4,5 and IFN-ү

M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081


44,45
LT, LT-B Similar to CT induce monocyte-derived Analogous structure and activities LT stimulates LCs cells, Rashes at local areas while applied as patch
DCs, fabrication of the CCR7, which to CT; improved uptake and rooting resettlement
is expressed on mature DC that habitat enhanced elicitation of response from skin to draining LNs.
to the lymph nodes and production of IL-12 to co-administered antigens;
produce ILs 2, 4, 5 and IFN-γ
46-48
LPS and its natural and LPS induces an IL-10-driven Induces migration of LCs; raises Along with polysaccharide-subunit Promising effects on neuro-inflammation
synthetic derivatives inhibition of CD4 T-cell T cell activation; improves the vaccine, developed Th1 and auto immune disorders being predicted.
(MPLA)/the outer expansion and has role in IL-10 degree and extent of immune responses to the carrier Strong prompter of pro-inflammatory cytokines
membrane production through monocytes responses; stimulates the host protein, with higher MPLA induce mild local reaction thus used as
of Gram-negative subsequent to attachment of cells to produce ILs and TNF. antibody response safe cutaneous adjuvant
bacteria PD-1 by PD-L1. LPS is the most in animal but in humans
Detoxified derivative potent endotoxin that induces
of LPS from production of tumor TNF-α by macrophages
Salmonella minnesota LPS and MPLA both are TLR-4 agonist
CpG ODN/bacterial CpG ODN binds with By improving the speed, degree, Phase 1 trials performed – 49,50

DNA and synthetic TLR9 located in endocytic time period and affinity of the in humans (in involvement
oligo-deoxy nucleotides vesicles. Unmethylated CpG defensive response, increases with Alum) have shown
motifs directly trigger an the vaccine's immunogenicity; improved anti-HBsAg
immuno-stimulatory promote production of antibody responses
surge that concludes in the Th1-driven response; generate Now oligonucleotides
maturation, differentiation, IFN-ү and proinflammatory classified into 3 classes
and propagation of multiple immune cytokines and the in consideration to their
cells, including B and T maturation/activation of distinct aptitude to trigger
lymphocytes, monocytes, DCs, professional APCs either: human B, NK or
NK cells and macrophages DCs in vitro.
Imiquimod Triggers skin immune cells via Convinces route of LCs; adapt Can stimulate DCs and B cells to – 51,52

TLR 7 and TLR 8 receptors, the immune response with induce Th 1 cell immunity, thus
also categorized as immune potent antiviral and anti-tumor boost up antibody production drained
modifiers, anti tumor possessions; a strong inducer out strong immune responses via microneedle
and antiviral effects of IFN-α, TNF-α delivery along with influenza subunit vaccine
and ILs GM-CSF more than vaccine alone in skin mediated
influenza vaccine
Cytokines: (IL-2, IL-12, Augmentation of – Flt3 along with HBV Exploitation of cytokines in cancer 49,53

GM-CSF, Flt3) antibody responses antigen induce the gathering of patients as Immuno adjuvant property
immature DCs in marginal blood, with some limitations such as some
without enhancement severe toxicity and
of antibody responses. short biological half-life.
CT: Vibrio cholerae cholera toxin; CT-B: B subunit of cholera toxin; LT: heat-labile enterotoxin of Escherichia coli; LT-B: heat-labile enterotoxin of E. coli B subunit; LPS: lipopolysaccharide,

1069
MPLA: monophosphoryl lipid A; IL: interleukin;
GM-CSF: granulocyte macrophage colony-stimulating factor; Flt3: fms-related tyrosine kinase 3 ligand.
1070 M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081

Figure 4. Schematic diagram of different approaches used to enhance skin permeation for TCI including physical, chemical, delivery systems and combination methods.

in concentrated amount on the patch surface might generate a vesicles. 105 It is well known that barrier of stratum corneum
high antigenic protein concentration gradient, which is signifi- prevents hydrophilic and large molecular drugs through skin but
cant for producing the driving force to hasten antigen transferosomes are capable to constrict through area 1/10th the
penetration. An extended immune response and booster effect width of the liposomes, permitting them to spontaneously
were also provoked indicating that the immune memory could be penetrate the stratum corneum. 106 Moreover, Cevc and
obtained by this patch-vaccine. 95 Glenn et al, 96 demonstrated Blume, 104 reported that the osmotic gradient which is an
that application of a patch containing heat labile enterotoxin (LT) influential force for their diffusion into the skin was the created
to humans has shown to produce robust LT-antibody responses. by the difference in water amount of the comparatively
These findings designated that TCI is feasible for human dehydrated skin surface and the hydrated viable epidermis.
immunization, and suggest that TCI may augment efficacy as Antigen entrapped in transferosomes induces stronger immune
well as improve vaccine delivery. Similarly, it was shown by response as compared to those entrapped in liposomes and
Ishii et al, 97 that patch based TCI system could induce effective present in antigenic solution alone. 107 Mishra et al, 72 reported
immune responses to viral and bacterial infection. Recently patch that transferosomes loaded with hepatitis B surface antigen
along with microneedles is used to provide non-invasive means (HBsAg) trigger enhanced antigen-specific systemic and muco-
of vaccination and further has been demonstrated by more and sal antibody titer responses against HBsAg in vivo by
more experiments and clinical trials. 98,99 subcutaneous administration than other formulations including
mixture of transferosomes and HBsAg, HBsAg solution alone
and alum-adsorbed HBsAg administered intramuscularly. Epi-
Second generation delivery systems (1990-2005) cutaneous immunization of transferosomes loaded with gap
Polymerosomes (PMs) junction proteins transcutaneously shows higher permeation and
relative higher anti-GJP IgA in the serum than conventional
PMs were first generation delivery system developed in late liposomes. 108
1990s. 100 They are self-assemble shells composed of amphi-
philic block copolymer, which consist of linked hydrophilic and Ethosomes
hydrophobic polymer chains and have amphiphilic properties Nowadays ethosomes have shown wide potential in TCI.
similar to phospholipids. 101 Polymerosomes have shown to be Safety concern and usefulness of ethosomes have been
stable in consideration of size and structure. 100 Based on the influentially established as compared to other transcutaneous
selection of the block copolymer, its physical properties, carriers such as gels, patches and conventional liposomes. 109
polymerosomes can be used as delivery systems with a wide They are vesicles composed of phospholipids, water and ethanol
range of suitable properties. 102 Furthermore, polymerosomes with a concentration range up to 45%. 110 Although, the exact
made up of degradable di-block polymer polyethylene glycol– mechanism for better permeation of the drugs into deeper layers
polybutadiene functionalized with an HIV-derived Tat peptide of the skin from ethosomes is still not clear. The synergistic
effectively deliver the PMs to the DCs. 103 These findings effects of two combinations are responsible for deeper
emphasize the potential exploit of polymerosomes as stout, virus distribution as well as penetration in the lipid bilayers of
like antigen delivery systems. skin. 111 The promising physical distinctiveness of ethosomes is
Transferosomes their softness, flexibility and deformability. Rattanapak et al, 112
in their studies concluded that for TCI cubosomes and ethosomes
Transferosomes, also referred as a new class of tailored are promising lipid carriers in contrast to transferosomes and
liposomes, were first reported by Cevc and Blume 104 and are liposomes. The first mechanism is the action of free ethanol
described as deformable, elastic or ultra-flexible liposomes or (ethanol effect). The polar head group of phospholipid present in
M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081 1071

Figure 5. Schematic diagram of several transcutaneous antigen delivery technologies for TCI (A) patch, (B) viral vectors, (C) sonophoresis, (D)
electroporation, (E) vesicular systems such as liposomes, transferosomes, niosomes, cubosomes etc, (F) micro or nano solutions and emulsions, (G) tape
stripping, (H) microneedles, (I) permeability enhancers, (J) cell penetrating proteins.

SC disrupts by free ethanol that causes the structures changes B for induction of humoral, cellular and mucosal immunity. 120
such as slackly disordered, increasing fluidity and membrane Their further research has shown that mucoadhesive ISCOMs
permeability. Then ethosomal vesicles (ethosomal effect), which that retain in nasal cavity for longer duration of time may reduce
are deformable and flexible straight forwardly infiltrate via the the dose/frequency for nasal vaccination. 121 Madsen and his
disorganized SC inside deeper into the skin. A further predicted coworkers 122 investigated the effect of novel type ISCOMs on
mechanism is the fusion between ethosomes and skin lipids, compounds skin penetration and their potential as delivery
ensuing in active moiety release from the vesicles. The system for TCI.
effectiveness of ethosomes for topical vaccine delivery has
Cubosomes
been explored. Mishra et al, 113 detailed that human DCs have
shown enhanced antigen uptake of HBsAg-loaded ethosomes Cubosomes are of the nanometric colloidal dispersion of
and the consequent triggering of an efficient Th1/Th2-type lipids having self assembling property that makes bicontinuous
immune response. cuboidal liquid crystalline particulate structures, with unique
practical properties able to encapsulate amphiphilic, hydrophilic
Immunostimulatory complexes (ISCOMs)
and hydrophobic active medicaments. 123 They have distinctive
Liposomes have strong potential for immunostimulation and enclosing, carrying, solubilizing and shielding properties
their uptake by DCs is rapid, but now they have largely been because of large surface area and low viscosity, making them
reinstated by the ISCOM formulations due to more stable an outstanding delivery system for various in vivo drug delivery
characteristics. ISCOMs are matrix constructs, incorporating routes. 124 Boyd 125 detailed that simple diffusion force resulting
antigen, cholesterol and phospholipids with a 40 nm diameter, a in rupture release of lipophilic drug from bulky cubic phases
size comparable to that of many viruses. 114,115 Liposomes are depends on size and molecular weight of the drugs. Cubosomes
readily incorporated into the membranes of cells, may promote have also been reported to act as a valuable vaccine delivery
endocytosis of antigen by DCs, monocytes and macrophages and system to enhance efficiency and reduce boosting requirement
can enter the endosomal pathway for MHC-Class II presentation with improved IFN-γ production in animals vaccinated subcu-
with DCs, but not other cell types. Thus, they efficiently promote taneously with ovalbumin and QA cubosomes as evaluated
T and B lymphoid cell activation. 116 Particles encapsulating against the control groups. 126 Bender et al, 127 envisaged about
antigens with a 30-200 nm diameter such as ISCOMs 117 and skin penetration of SRB, a fluorescence hydrophilic model drug
virus like particles (VLPs) usually also provide “threat” signals encapsulated in cubosomes of mono-olein with the two-photon
that induce activation of APCs via TLRs. 118 Pandey et al, 119 microscopy and high accumulation fluroscence intensity in
prepared ISCOMs incorporating rHBsAg when administered micro-fissures and from which threadlike structure of fluores-
subcutaneously were shown to induce humoral as well as cellular cence arrangement expands out laterally in skin tissues.
immunity and found to be immunogenic as compared to Vesosomes
marketed rHBsAg formulations by subcutaneous route of
administration. They also demonstrated potential of ISCOMs Vesosomes are multicompartment structures which have
as a carrier adjuvant for nasal subunit vaccines against hepatitis distinct inner compartments separated from the external
1072 M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081

Table 4
Different generation delivery systems used for transcutaneous immunization.
Particulate Principle Properties Limitations Animal/effects Reference
systems
First generation delivery systems (before 90s)
67-71
Liposomes Inter membrane transfer, Enhance activity against Biocompatible and Mice/induce IgG specific
adsorption extra and intracellular ecofriendly, low antibody, elicit Th1 and
Fusion, phagocytosis pathogens, effective efficiency rapid clearance Th2 specific antibody response
immunoadjuvant from blood stream by
properties, enhance phagocytic cells
penetration
73-76,80,88
Nanoparticles Disruption of SC Seems alike natural pathogen in Can lead to particle Mice/increase uptake of
by nano-bio interaction size and shape actively taken aggregation due to small antigen by LCs and increase
with skin lipids up by APCs improve antigen size and large surface level of IFN-γ and IL-2.
uptake and immunogenicity area, limited loading,
sometimes toxic
product form
89,90,92,94
Niosomes Diffusion, interaction Have characteristic size, shape May undergo fuse, Mice/elicit strong IgG
with SC via fusion, and fluidity, can entrap wide aggregate, leaking antibody titers, stimulate
aggregation and adhesion range of molecules of entrapped serum immune responses
drugs, hydrolysis
95-97
Patches Present high Ag protein Promotes Ag penetration Local irritation and Rat/induce mainly
concentration gradient which across SC, induce Ag sensitization, Th2 type immune response
produce high driven force specific immune response contact dermatitis and antigen specific
for Ag penetration immune memory
Second generation delivery system (1999-2005)
100-103
Polymerosomes Internalization by Self assemble shell, amphiphilic Low transfection efficiency, Mice/effective
endocytosis, hydrolytic properties make them suitable short-lived expression deliver antigen to DCs
degradation carrier for both type molecules, of foreign gene
greater stability
104-107,72,108
Transferosomes Via diffusion Ultra flexibility, induce stronger Chemically unstable, Mice/robust mucosal and
due to osmotic immune response Compared inability to transport large systemic antibody response
gradient force to conventional liposomes molecules, expensive
109,111-113
Ethosomes Fusion with cell Via increasing cell membrane Low diffusion Mice/enhanced
membrane proteins fluidity increase skin penetration, of Ag across SC Th1/Th2 cytokines
improved Ag entrapment efficiency level and uptake by DCs
114-116,119,122
ISCOMs Endocytosis by APCs Strong immuno-stimulation High cost Mice/upregulation of
potential, promote T and B Quil A toxicity humoral (IgG) as well as
cells activation cellular (IFN-γ and
IL-2) levels
124-127
Cubosomes Simple diffusion Distinctive enclosing, Low loading efficiency Mice/higher IFN-γ
rapture release carrying, solubilizing, and less stability level and high penetration
shielding properties of peptide in skin
128-130,130
Vesosomes Fusion with target cells Have multiple Difficult to encapsulate Rat/significantly induce
compartment, can Large molecules serum IgG titer
encapsulate multiple and Th1 and Th2
material in same structure combined response
Third generation delivery system (2005 to present)
132,133,136,137
Gold NPs Via membrane interaction Huge surface area, high Cytotoxicity Mice/increased level
and penetration without stability, easily fabricable of antigen specific
membrane disruption in different shape IgG and deep
penetration in skin layers

membrane. In a straightforward way it can be said as a larger the vesosomes, while small molecules are released from
vesicle that deliberately encapsulates many smaller size vesicles unilamellar liposomes in minutes, they are retained in vesosomes
inside it. Each compartment of vesosome can encapsulate from hours to days, even though the liposomes and vesosomes
different materials and have different bilayer composition. 128,129 have the same bilayer composition and size. 131 Mishra et al, 130
Vesosomes (fusion liposomes) are novel carriers that can have demonstrated the fusion ability properties of phospholipid
competently support the intracellular delivery of encapsulated liposomes made up of phosphatidylcholine dioleoyl phosphati-
antigen by fusion with target cells. The main drawback of dylethanolamine (DOPE), dioleoyl trimethyl ammonium pro-
liposomes is that various vital drugs are released earlier than pane (DOTAP) and dipalmitoyl phosphatidylcholine (DPPC) to
favorable in vivo. 130 This problem is significantly addressed by combine with principal phospholipid containing membrane
M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081 1073

Figure 6. Schematic diagram showing generation of adaptive immune response following topical application of antigen containing NPs with or without adjuvant.

constituent. These types of fusogenic vesosomes have ability to are reviewed in the next section and relevant review articles are
fuse with membrane of target cells and elicited strong immune cited to provide further details.
response after transcutaneous vaccination.
Gold NPs (AuNPs)

Relevance of gold NPs (AuNPs) in biomedical and bioima-


Third generation delivery systems (2005-present) ging constitutes a fast-growing field. 135 An interesting recent
Inorganic nanoparticulate based vaccine-codelivery nanocar- report was revealed that superfine Au-NPs 5 nm were
rier investigated for TCI due to their skin permeable property.
Huang et al, 136 reported that when model protein drugs such as
Metal NPs such as quantum dots and iron oxide NPs are able horseradish peroxidase (HRP), β-galactosidase (β-gal) and
to penetrate through intact skin, of less than 10 nm in size, due to ovalbumin (OVA) were co-administered with Au-NPs (5 nm)
its interaction with the lipid in extracellular space. Biocompatible their dermal absorption increases and migrates into the deep skin
inorganic nanoparticulate based vaccine-codelivery is a new layers, which successfully enhanced the dermal absorption of
choice that successfully evades the basic drawbacks of model proteins. It should be renowned that before application of
conventional organic material carrier system in nanomedicine, formulation skin hydration had been done for 10 min by covering
biomolecular and cellular applications, due to its inimitable with the help of gauze to smoothen the process of uptake. In
structural and compositional characteristics, for example huge another experiments cholera toxin (CT), a strong mucosal
surface area, tunable surface characteristics by engineering adjuvant along with OVA was co-applied with the Au particles.
modifications and surface functionalization, plentiful physio- Normal saline solution of CT applied on back surface of shaved
chemical functionalities, high stability and explicit biological mouse may play a role of an adjuvant for co-administered
nature. 132,133 Advancements in inorganic nanocarrier develop- vaccine and provoke high altitudes of CT-specific IgG
ment have made up our mind to focus on inorganic NPs for antibodies. Cholera toxin is also known for its leakable effect.
controlled release specific targeting and control of their cellular Au-NPs accumulated in high amounts in the stratum corneum
actions. 134 Yet much of work on inorganic NPs for TCI has not and in the epidermis. OVA along with CT in Au-NPs elicited the
been pursued. Elected applications of some useful inorganic NPs production of IgG. 137 Recently, functionalized AuNPs have
1074 M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081

Table 5
Recent clinical trial studies on transcutaneous immunization.
Title Clinical trial no. Clinical trial initiation and completion date Status Reference
181
ETEC Logical Trial (TREK) NCT00516659 May 2006/December 2007 Phase II (Completed)
182
Transcutaneous Immunization With An Attenuated NCT01311817 January 2011/July 2012 Phase I (Completed)
Listeria Monocytogenes Vector Vaccine
183
Safety Study of Recombinant Vaccine NCT01382095 July 2011/April 2013 Phase I (Completed)
to Prevent ETEC Diarrhea
184
Safety Study of Chimeric Vaccine to NCT01644565 August 2012/November 2016 Phase I (Ongoing)
Prevent ETEC Diarrhea (Expected completion)
185
Traveler's Diarrhea (TD) Automated Process NCT00751777 September 2008/November 2009 Phase II (Completed)
186
LT Vaccine Patch Self-Administration Study NCT00565461 November 2007/August 2008 Phase II (Completed)
187
Gender-Stratified Safety and Immunogenicity Study NCT01067781 February 2010/March 2012 Phase II (Completed)
188
Study of Allergen Immunotherapy in Grass NCT00777374 October 2008/November 2010 Phase II (Completed)
Pollen Allergic Subjects With Epicutaneous
Allergen (ZU-SkinSIT-003)
189
Safety Study Comparing a Vaccine NCT00261001 October 2005/September 2009 Phase I (Completed)
Transcutaneous Administration to
the Intramuscular Route (MANON-05)
190
Immunogenicity of Three NCT02075983 June 2013/December 2015 Phase I (Ongoing)
HIV GTU® Multi HIV DNA (Expected completion)
Immunizations Administered
Via Intramuscular, Intradermal
and Transcutaneous routes
(CUT‫٭‬HIVAC001)
191
Routes of Immunization And NCT01707602 October 2012/April 2013 Phase I/II (Completed)
Flu Immune Response
192
The safety immunogenicity of the DNA-GTU NCT02457689 May 2015/February 2017 Phase I/II (Completed)
Vaccine Administered to HIV-infected patients
on ART vs. Placebo (CUTHIVTHER001)
193
Vaccine Therapy and Resiquimod in Treating NCT00470379 April 2006/October 2012 Phase 0 (Completed)
Patients With Stage II, Stage III OR Stage
IV Melanoma That Has Been
completely removed by surgery
194
Immune response to yellow fever vaccination NCT00723489 August 2008/April 2011 Phase I/II (Completed)
in adults with atopic dermatitis
195
Study of safety and immunogenicity ISRCTN88861431 March 2004/March 2005 Phase I/II (Completed)
of Measles vaccine (Rouvax®)
administered by transcutaneous route

emerged as a delivery system for targeting, bioimaging and enhancer via different mechanism of action to overcome skin
biodistribution studies, 138 -141 biomolecules 142 or large size barrier and show synergistic effects action in TCI. Examples
biomolecules such as proteins, 143 DNA, 144 and RNA. 145 utilizing these approaches are below.

Synergistic enhancers/system combinations


Combination methods It have been observed that the majority of the enhancers that
Complex formulations have been regarded as in the transdermal literature for their
ability to increase transport across skin have important
In previous text we had discussed about various delivery limitations such as not able to achieve the desired skin disruption
systems, physical approaches and chemical approaches to and cause skin irritation that is associated with their potency of
overcome the SC for TCI. But to be applicable in clinical use penetration. 146,147 Chemical mixtures of these enhancers or
almost it's a failure to achieve this goal with the use of single synergistic combinations present several possible opportunities
approaches we discussed above. So it's a prerequisite to use to surmount the restrictions of single chemical enhancers. Many
either combination of delivery systems together or delivery examples of classical combinations are available such as
systems with chemical approaches or with physical approaches chemical mixture of two chemical enhancers, one of them
or all the three approaches together. They provide quite better works on keratinocytes to open up transcellular and intracellular
results in combination than they used alone. For example, when pathways for infiltration of active moieties molecules and other
two delivery systems used together for TCI suppose liposomes one acts on lipids of skin. Likewise, one component of mixture
with NPs, here liposomes containing lipid so they work to disrupt combination of enhancer increases diffusion of therapeutic
SC and itself as adjuvant and NPs used as carrier system to molecules in stratum corneum and other one creates pathways for
deliver vaccine material. Similar results have shown by chemical them via disrupting. Some chemical enhancers in this
M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081 1075
157 158
combinations are added to decrease or prevent degradation or to well as cellular immunity. Makidon et al, who prepared
enhance stabilization of therapeutics because they have proper- nanoemulsion adjuvant system of rHBsAg for nasal vaccination
ties of antioxidant, stabilizer and prevent enzymatic degradation demonstrated that this vaccine induces a Th1 associated cellular
of proteins, peptides and vaccines. 148,149 immunity. Supplementary research has been ongoing in animal
trials for other vaccines including anthrax and hepatitis B. This
Biphasic vesicles nanoemulsion technology has been licensed by NanoBio
Corporation for vaccine development. 159
Lipid based delivery systems which are composed of both
liposomes and emulsions are called biphasic systems. Biphasic
vesicle systems provide several advantages as a topical carrier Other formulations
system such as having multichamber structure allows encapsu-
lation and co-encapsulation of various therapeutics of different Other than above mentioned methods and delivery systems,
weight range with different solubilities. Biphasic systems are more new approaches to overcome problems associated with TCI
evaluated for many vaccine candidates including proteins, have been discussed here. Su et al, 160 reported about
nucleotides, recombinant subunits and classical old inactivated hydrolytically degradable loaded protein and oligonucleotide
or killed vaccines and now became applicable to purposes in was incorporated into a novel multilayer film for the transcu-
human as well as in animals. 150 -152 Baca-Estrada et al, 150 have taneous delivery. Results showed OVA loaded from LbL films
shown topical delivery of hen egg lysozyme (HEL) and penetrated into barrier-disrupted skin was uptaken by skin APCs
leukotoxin antigens in biphasic delivery system induce strong and transported to the skin-draining lymph nodes. Tahara et
Th-2 cellular response and induce secretion of IgG1 antibody al, 161 successfully achieved TCI by applying a solid-in-oil (S/O)
response. Foldveri et al, 151 formulated biphasic delivery system nanodispersion, without the use of any skin pre-treatment or
of plasmid DNA (pDNA) and have shown that it delivered adjuvant. S/O of hydrophilic drugs has effectively enhanced the
significant quantities of pDNA into the viable layers of human permeation of proteins into the skin. Result outcomes have
skin in vitro and induced 5 times higher anti-IgD levels shown that upon incorporation of low molecular drug in elastic
compared with naked DNA alone and secreted IL-4 by cells vesicles enhanced their transdermal diffusion through the skin
and induced cellular immune response. Babiuk et al, 152 and vesicular structures are reached in the deep layer of stratum
demonstrated that topical delivery of plasmid in biphasic lipid corneum. In another report about, nanofiber merged, an
delivery system results to induce cellular and humoral response. uninterrupted, nonwoven nanofibrous membrane containing
Intradermal application induces Th-1 whereas TCI induces Th-2 nanoencapsulated protective antigen (PA) is produced through
type cellular immune response. the process of electrospinning. This novel formulation provides
the basis for the transdermal anthrax vaccine. 162
Micro and nanoemulsions

Micro and nanoemulsions is colloidal carrier system that Devices and formulations
offers several advantages for drug delivery, such as ease of As we previously discussed in this review removal or
preparation, long-standing stability, soaring solubilization ca- disruption of SC increases the transcutaneous permeability for
pacity for hydrophilic and lipophilic agents. 153 Microemulsion antigens and makes them effectively available for captured by
has shown great prospective in absorption and delivery APCs present in skin layers. Many studies also revealed that
enhancement of peptides and pDNA, especially the water-in-oil transcutaneous application of formulation system alone was not
(W/O) type, 154 and shown enhanced penetration properties in able to induce effective immune responses. To make these
stratum corneum due to their lipophilic nature. 155 Although it approaches possible physical methods and chemical enhancers
has not been widely reported yet for the application in along with novel vaccine delivery systems are used. Here below
transcutaneous vaccine, nanoemulsion has showed its advantage we shortly discussed about this concept with few examples.
in topical delivery of hydrophobic drugs due to their lipophilic
nature. Nanoemulsions are isotropic novel drug delivery systems Chemical enhancers with delivery systems
consisting of two liquid phases immiscible with each other,
emulsified oil and water systems with mean nanometric droplet Synergistic combination possible for vesicular colloidal
size range. They are either oil-in-water (o/w) or water-in-oil (w/ delivery is made up of concentric bilayers of self-assembled
o) type systems, where the internal phase of the particle is either amphiphilic molecules. Vesicular systems have achieved
oil or water, respectively. Nanoemulsion containing antigen and eminence as skin penetration improving agents as well as
adjuvants effectively taken up by APCs efficiently induces protein/vaccine carrier agents in transdermal delivery. 69,163 The
cellular as well as humoral immunity and increases secretion of physiochemical characteristics of chemical enhancers have an
IgG and IgA antibodies in serum. So emergent mucosal intense outcome on the deeds of the vesicles and therefore on
immunity via use of nanoemulsion may turn out to be an their efficiency in enhancing vaccines and therapeutics delivery
important tool in the future struggle against HIV. 156 In some through skin. 104,112,163-169 Synergistic relations between the
cases, nanoemulsion works as adjuvant for vaccine formulations components of the vesicular system and between the different
applied to the mucosal surface and it helps uptake by APCs. This vesicular systems and skin elements are alleged to be
results in the significant systemic and mucosal immune response accountable for the advanced skin permeation enhancement
due to that the production of specific IgG and IgA antibody as through vesicular systems.
1076 M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081

Physical methods with delivery systems Many diverse advances have been enlarged of which quite a lot
of methods may escort to flourishing TCI by combining the
Various physical methods are available to increase TCI novel formulation approaches with physical/chemical ap-
potential such as microneedle techniques, ultrasound, electropo- proaches together with addition of an adjuvant that activates
ration, laser, hyperthermia technique, gene gun and tape specific receptor for robust immune response which we have
stripping etc. microneedle array mostly prepared along with discussed in our section approaches to improve TCI. As we
patch formulations and being delivered to the skin in painlessly discussed in this review, various approaches to disrupt stratum
manner. Alarcon et al, 170 reported that influenza vaccine corneum and formulations have been developed, further, basic,
administration along with microneedle induces higher immune clinical and preclinical studies have also been conducted, but still
response than that of intramuscular injection in animal model. most of transcutaneous vaccines are in preclinical or clinical trial
Koutsonanos et al via metal microneedle deliver inactivated stage just because the most efficient way toward this is at a halt
influenza lethal virus and induce strong immune response. 171 and desires to be established, which will necessitate cooperative
Similar finding for immunization by vaccine coated micro- endeavors of pharmaceutical experts, vaccine development
needles is shown in several studies. 98,99,171,172 Tape stripping experts, immune experts and mechanical engineer experts.
and degreasing with chemical promote dermal immunization via Only then TCI can effectively improve and in the near future
skin disorganization, partial leaching of lipids and induce APCs revolutionize vaccination method which would contribute to a
maturation and migration. 173,174 Huang et al, 175 using low global countermeasure against infectious diseases and would
pressure gene gun deliver NPs transcutaneously and induce provide many countries with cost effective and beneficial
strong Th2 immune response. Electroporation high voltage pulse vaccination.
techniques cause structural perturbation of skin lipid bilayer and
are widely used in TCI. 176 Low frequency ultrasound techniques
References
increase antibody titer because of their adjuvant action and are
used in delivery of vaccine delivery systems through induced
inflammation. 177,178 Thermal hyperthermia through activation
1. Hilleman MR. Vaccines in historic evolution and perspective: a
and migration of LCs and because not causing any pain and narrative of vaccine discoveries. Vaccine 2000;18:1436-47.
damage to skin should be promoted as a unique technique for 2. Schijns VE. Immunological concepts of vaccine adjuvant activity. Curr
TCI. 179,180 All the above examples have shown that physical Opin Immunol 2000;12:456-63.
method along with delivery system came to be beneficial for 3. Raz E, Carson DA, Parker SE, Parr TB, Abai AM, Aichinger G, et al.
improved TCI. Intradermal gene immunization: the possible role of DNA uptake in the
induction of cellular immunity to viruses. Proc Natl Acad Sci U S A
1994;91:9519-23.
Clinical trials of transcutaneous immunization
4. Combadiere B, Liard C. Transcutaneous and intradermal vaccination.
Hum Vaccine 2011;7:811-27.
There are several numbers of diseases such as tetanus,
5. Jacobson RM, Swan A, Adegbenro A, Ludington SL, Wollan PC,
measles, mumps, pertussis, rubella, diphtheria, Haemophilus Poland GA, et al. Making vaccines more acceptable—methods to
influenzae type b (Hib), yellow fever and polio which are from prevent and minimize pain and other common adverse events
very last decade under command because of vaccine creation. associated with vaccines. Vaccine 2001;19:2418-27.
The attention on the development of vaccine becomes more 6. Sumida SM, McKay PF, Truitt DM, Kishko MG, Arthur JC, Seaman
attractive due to the wide use of TCI. Several preclinical trials of MS, et al. Recruitment and expansion of dendritic cells in vivo
transcutaneous immunization have been reported and some of potentiate the immunogenicity of plasmid DNA vaccines. J Clin Invest
2004;114:1334-42.
them successfully reached the clinical trial stage. Clinical trials
7. Torres CA, Iwasaki A, Barber BH, Robinson HL. Differential
reported on TCI in recent few years are summarized in Table 5. dependence on target site tissue for gene gun and intramuscular DNA
immunizations. J Immunol 1997;158:4529-32.
Conclusions and future prospective 8. Stan AC, Casares S, Brumeanu TD, Klinman DM, Bona CA. CpG
motifs of DNA vaccines induce the expression of chemokines and
The prime motive of vaccine development is the establish- MHC Class II molecules on myocytes. Eur J Immunol 2001;31:301-10.
ment of manufacturing technologies that provide secure and 9. Iwasaki A, Stiernholm BJ, Chan A, Berinstein NL, Barber BH.
effectual vaccine antigen in haste and soundly, but this problem Enhanced CTL responses mediated by plasmid DNA immunogens
to bear out as much as necessary vaccination to prevent encoding co stimulatory molecules and cytokines. J Immunol
1997;158:4591-601.
infectious diseases stays behind to be solved. It is now a
10. Simerska P, Moyle PM, Olive C, Toth I. Oral vaccine delivery—new
well-known fact that the skin is an attractive immunological strategies and technologies. Curr Drug Deliv 2009;6:347-58.
avenue that offers non-invasive, needle free, painless, effective, 11. Slutter B, Hagenaars N, Jiskoot W. Rational design of nasal vaccines. J
easy-to-use, low cost prophylaxis against infectious diseases, and Drug Target 2008;16:1-17.
better patient compliance with few side effects and secured 12. Mikszta JA, Laurent PE. Cutaneous delivery of prophylactic and
handling than conventional injections. The main challenges to be therapeutic vaccines: historical perspective and future outlook. Expert
Rev Vaccines 2008;7:1329-39.
pointed in transcutaneous vaccination are to make certain precise
13. Lu D, Hickey AJ. Pulmonary vaccine delivery. Expert Rev Vaccines
delivery into the epidermal or dermal skin of antigens through 2007;6:213-26.
subcutaneous where LCs and DCs exist and to formulate antigen 14. Ryan EJ, Daly LM, Mills KH. Immunomodulators and delivery
with adjuvant and/or carrier system for effectual activation of systems for vaccination by mucosal routes. Trends Biotechnol
defined type of cells/receptors/ligands that exists in DC subsets. 2001;19:293-304.
M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081 1077

15. Amorij JP, Kersten GF, SalujaV TonnisWF, Hinrichs WL, Slutter B, et al. 40. Gupta RK, Siber GR. Comparison of adjuvant activities of aluminium
Towards tailored vaccine delivery: needs, challenges and perspectives. phosphate, calcium phosphate and stearyl tyrosine for tetanus toxoid.
J Control Release 2012;161:363-76. Biologicals 1994;22:53-63.
16. Kupper TS, Fuhlbrigge RC. Immune surveillance in the skin: mechanisms 41. Manning BM, Enioutina EY, Visic DM, Knudson AD, Daynes RA.
and clinical consequences. Nat Rev Immunol 2004;4:211-22. CpG DNA functions as an effective adjuvant for the induction of
17. Banchereau J, Steinman RM. Dendritic cells and the control of immune responses in aged mice. Exp Gerontol 2001;37:107-26.
immunity. Nature 1998;392:245-52. 42. Seya T, Akazawa T, Tsujita T, Matsumoto M. Role of Toll-like
18. Bal SM, Ding Z, van Riet E, Jiskoot W, Bouwstra JA. Advances in receptors in adjuvant-augmented immune therapies. Evid Based
transcutaneous vaccine delivery: do all ways lead to Rome? J Control Complement Alternat Med 2006;3:31-8.
Release 2010;148:266-82. 43. Anjuere F, George-Chandy A, Audant F, Rousseau D, Holmgren J,
19. Seneschal J, Jiang X, Kupper TS. Langerin + Dermal DC, but not Czerkinsky C. Transcutaneous immunization with cholera toxin B
Langerhans cells, are required for effective CD8 mediated immune subunit adjuvant suppresses IgE antibody responses via selective
responses after skin scarification with Vaccinia Virus (VACV). J Invest induction of Th1 immune responses. J Immunol 2003;170:1586-92.
Dermatol 2014;134:686-94. 44. Choi AH, Smiley K, Basu M. Induction of immune responses and
20. Ponvert C, Scheinmann P. Vaccine allergy and pseudo-allergy. Eur J partial protection in mice after skin immunization with rotavirus VP6
Dermatol 2003;13:10-5. protein and the adjuvant LT(R192G). Vaccine 2005;23:2290-3.
21. Rios M. Advances in transdermal technologies: transdermal delivery 45. Pitcovski J, Bazak Z, Wasserman E, Elias O, Levy A, Peretz T, et al.
takes up once forbidden compounds, reviving markets and creating Heat labile enterotoxin of E. coli: a potential adjuvant for transcuta-
formulation opportunities. Pharma Technol 2007:54-8. neous cancer immunotherapy. Vaccine 2006;24:636-43.
22. Gordon Ryan D. More than skin deep: advances in transdermal 46. Said EA, Dupuy FP, Trautmann L, Zhang Y, Shi Y, El-Far M, et al.
technologies are opening up new avenues of exploration. Pharm Programmed death-1-induced interleukin-10 production by monocytes
Technol Eur 2005;17:66. impairs CD4 + T cell activation during HIV infection. Nat Med
23. Matzinger P. The danger model: a renewed sense of self. Science 2010;16:452-9.
2002;296:301-5. 47. Raina N, Matsui J, Jeejeebhoy KN. Nutritional and metabolic effects of
24. Asahina A, Tamaki K. Role of Langerhans cells in cutaneous protective the endotoxin bacterial lipopolysaccharide in orally and parenterally fed
immunity: Is the reappraisal necessary? J Dermatol Sci 2006;44:1-9. rats. Am J Clin Nutr 2000;71:835-43.
25. Ludwig IS, Geijtenbeek TB, Van Kooyk Y. Two way communication 48. Chen X, Wu MX. Laser vaccine adjuvant for cutaneous immunization.
between neutrophils and dendritic cells. Curr Opin Pharmacol Expert Rev Vaccines 2011;10(10):1397-403.
2006;6:408-13. 49. Hickey DK, Aldwell FE, Tan ZY, Bao S, Beagley KW. Transcutaneous
26. Demento SL. Inflammasome-activating NPs as modular systems for immunization with novel lipid-based adjuvants induces protection against
optimizing vaccine efficacy. Vaccine 2009;27:3013-21. gastric Helicobacter pylori infection. Vaccine 2009;27:6983-90.
27. Teunissen MB, Kapsenberg ML, Dos JD. Langerhans cells and related 50. Hickey DK, Aldwell FE, Beagley KW. Transcutaneous immunization
skin dendritic cells: skin immune system. 2nd ed. CRC, Press LLC; 1997. with a novel lipid-based adjuvant protects against Chlamydia genital
28. Williams A, Richard AF, Stephanie CE. The role of NOD-like receptors and respiratory infections. Vaccine 2009;27:6217-25.
in shaping adaptive immunity. Curr Opin Immunol 2010;22:1-7. 51. Johnston D, Bystryn JC. Topical imiquimod is a potent adjuvant to a
29. Medzhitov R, Preston-Hurlburt P, Janeway Jr CA. A human weakly-immunogenic protein prototype vaccine. Vaccine
homologue of the Drosophila Toll protein signals activation of adaptive 2006;24:1958-65.
immunity. Nature 1997;388:394-7. 52. Zhu J, Lai K, Brownile R, Babiuk LA, Mutwiri GK. Porcine TLR8 and
30. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate TLR7 are both activated by a selective TLR7 ligand, imiquimod. Mol
immunity. Cell 2006;124:783-801. Immunol 2008;45:3238-43.
31. Lauw FN, caffery DR, Golenbock DT. Of mice and man: TLR11 53. Hickey DK, Bao S, Ikeda LT, Carey AJ, Beagley KW. Induction of
(finally) finds profilin. Trends Immunol 2005;26:509-11. antichlamydial mucosal immunity by transcutaneous immunization is
32. Heil F, Ahmad-Nejad P, Hemmi H, Hochrein H, Ampenberger F, enhanced by topical application of GM-CSF. Curr Mol Med
Gellert T, et al. The Toll-like receptor 7 (TLR7)-specific stimulus 2005;5:559-605.
loxoribine uncovers a strong relationship within the TLR7, 8 and 9 54. kim YC, Prausnitz MR. Enabling skin vaccination using new delivery
subfamily. Eur J Immunol 2003;33:2987-97. technologies. Curr Top Microbiol Immunol 2012;351:77-112.
33. Matsumoto M, Funami K, Tanabe M, Oshiumi H, Shingai M, Seto Y, et 55. Sullivan SP, Koutsonanos DG, Del Pilar Martin M, Lee JW, Zarnitsyn
al. Subcellular localization of Toll-like receptor 3 in human dendritic V, Choi SO, et al. Dissolving polymer microneedle patches for
cells. J Immunol 2003;171:3154-62. influenza vaccination. Nat Med 2010;16:915-20.
34. Van der Aar AM, Sylva-Steenland RM, Bos JD, Kapsenberg ML, de 56. Matsuo K, Hirobe S, Yokota Y, Ayabe Y, Seto M, Quan YS, et al.
Jong EC, Teunissen MB. Loss of TLR2, TLR4, and TLR5 on Transcutaneous immunization using a dissolving microneedle array
Langerhans cells abolishes bacterial recognition. J Immunol protects against tetanus, diphtheria, malaria, and influenza. J Control
2007;178:1986-90. Release 2012;160:495-501.
35. Kelsoe G. Studies of the humoral immune response. Immunol Res 57. Skountzou I, Quan FS, Jacob J, Compans RW, Kang SM.
2000;22:199-210. Transcutaneous immunization with inactivated influenza virus induces
36. Li N, Peng LH, Chen X, Nakagawa S, Gao JQ. Transcutaneous protective immune responses. Vaccine 2006;24:6110-9.
vaccines: novel advances in technology and delivery for overcoming 58. Glenn GM, Kenney RT, Ellingsworth LR, Frech SA, Hammond SA,
the barriers. Vaccine 2011;29:6179-90. Zoeteweij JP. Transcutaneous immunization and immunostimulant
37. Van der Laan JW. Adjuvants enhancing an integral immune response to strategies: capitalizing on the immunocompetence of the skin. Expert
antigens. Expert Rev Vaccines 2005;4:15-8. Rev Vaccines 2003;2:253-67.
38. Himes R, Lee S, McMenigall K, Russell-Jones G. The influence of 59. Singh D, Pradhan M, Nag M, Singh MR. Vesicular system: versatile
molecular adjuvants in the cutaneous response to antigen after topical carrier for transdermal delivery of bioactives. Artif Cells Nanomed
vaccination. Vaccine 2011;29:5393-8. Biotechnol 2014;25:1-9.
39. Tomoka TN, Erika T, Megumi K, Koichi T. Transcutaneous 60. Songkro S. An overview of skin penetration enhancers: penetration
immunization system using a hydrotropic formulation induces a potent enhancing activity, skin irritation potential and mechanism of action.
antigen-specific antibody response. PLoS One 2010;7:e47980. Songklanakarin J Sci Technol 2009;31:299-321.
1078 M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081

61. Parhi R, Suresh P, Mondal S, Kumar PM. Novel penetration enhancers 83. Nagpal K, Singh SK, Mishra DN. Chitosan NPs: a promising
for skin applications: a review. Curr Drug Deliv 2012;9:219-30. system in novel drug delivery. Chem Pharm Bull (Tokyo)
62. Scharton-Kersten T, Glenn GM, Vassell R, Yu J, Walwender D, Alving 2010;58:1423-30.
CR. Principles of transcutaneous immunization using cholera toxin as 84. Prego C, Paolicelli P, Diaz B, Vicente S, Sanchez A, Gonzalez-
an adjuvant. Vaccine 1999;17:S37-43. Fernandez A, et al. Chitosan-based NPs for improving immunization
63. Karande P, Arora A, Pham TK, Stevens D, Wojicki A, Mitragotri S. against hepatitis B infection. Vaccine 2010;28:2607-14.
Transcutaneous immunization using common chemicals. J Control 85. He W, Guo X, Zhang M. Transdermal permeation enhancement of N-
Release 2009;138:134-40. trimethyl chitosan for testosterone. Int J Pharm 2008;356:82-7.
64. Kogan A, Garti N. Microemulsions as transdermal drug delivery 86. Slutter B, Plapied L, Fievez V, Sande MA, des Rieux A, Schneider YJ,
vehicles. Adv Colloid Interface Sci 2006;123–126:369-85. et al. Mechanistic study of the adjuvant effect of biodegradable NPs in
65. Touitou E, Godin B. Enhancement in drug delivery. In: Touitou E, mucosal vaccination. J Control Release 2009;138:113-21.
Barry B, editors. Boca Raton FL: CRC Press; 2007. p. 255-78. 87. Bal SM, Slutter B, Jiskoot W, Bouwstra JA. Small is beautiful: N-
66. Babiuk S, Baca-Estrada M, Babiuk LA, Ewen C, Foldvari M. trimethyl chitosan-ovalbumin conjugates for microneedle-based trans-
Cutaneous vaccination: the skin as an immunologically active tissue cutaneous immunisation. Vaccine 2011;29:4025-32.
and the challenge of antigen delivery. J Control Release 88. Li N, Peng LH, Chen X, Zhang TY, Shao GF, Liang WQ, et al.
2000;66:199-214. Antigen-loaded nanocarriers enhance the migration of stimulated
67. Combadiere B, Mahe B. Particle-based vaccines for transcutaneous Langerhans cells to draining lymph nodes and induce effective
vaccination. Comp Immunol Microbiol Infect Dis 2008;31:293-315. transcutaneous immunization. Nanomedicine 2014;10:215-23.
68. Castro GA, Ferreira LA. Novel vesicular and particulate drug delivery 89. Jain CP, Vyas SP, Dixit VK. Niosomal system for delivery of
systems for topical treatment of acne. Expert Opin Drug Deliv rifampicin to lymphatics. Indian J Pharm Sci 2006;68:575-8.
2008;5:665-79. 90. Khan A, Sharma PK, Visht S, Malviya R. Niosomes as colloidal drug
69. Cosco D, Celia C, Cilurzo F, Trapasso E, Paolino D. Colloidal carriers delivery system: a review. J Chronother Drug Deliv 2011;2:15-21.
for the enhanced delivery through the skin. Expert Opin Drug Deliv 91. Jain S, Vyas SP. Mannosylated niosomes as carrier adjuvant system for
2008;5:737-55. topical immunization. J Pharm Pharmacol 2005;57:1177-84.
70. Elsayed MMA, Abdullah OY, Naggar VF, Khalafallah NM. Deform- 92. Maheshwari C, Pandey RS, Chaurasiya A, Kumar A, Selvam DT,
able liposome and ethosome: mechanism of enhanced skin delivery. Int Prasad GBKS, et al. Non-ionic surfactant vesicles mediated transcu-
J Pharm 2006;322:60-6. taneous immunization against hepatitis B. Int Immunopharmacol
71. Tyagi RK, Garg NK, Jadon R, Sahud T, Katare OP, Dalai SK, et al. 2011;11:1516-22.
elastic liposome mediated transdermal immunization enhanced the 93. den Bergh BA Van, Frederik PMF, Bomans PHH, Junginger HE,
immunogenicity of P. falciparum surface antigen MSP-119. Vaccine Bouwstra AJ. Elastic liquid state vesicles as a tool for topical drug
2015;33:4630-8. delivery. Division of Pharmaceutical Technology, Leiden University,
72. Mishra D, Dubey V, Asthana A, Saraf DK, Jain NK. Elastic liposomes The Netherlands; 1999. p. 49-63.
mediated transcutaneous immunization against hepatitis B. Vaccine 94. Ding Z, Bivas-Benita M, Hirschberg H, Kersten GF, Jiskoot W,
2006;24:4847-55. Bouwstra JA. Preparation and characterization of diphtheria toxoid-
73. Perrie Y, Mohammed AR, Kirby DJ, McNeil SE, Bramwell VW. loaded elastic vesicles for transcutaneous immunization. J Drug Target
Vaccine adjuvant systems: enhancing the efficacy of sub-unit protein 2008;16:555-63.
antigens. Int J Pharm 2008;364:272-80. 95. Matsuo K, Ishii Y, Quan YS, Kamiyama F, Mukai Y, Yoshioka Y, et al.
74. Schlosser E, Mueller M, Fischer S, Basta S, Busch DH, Gander B, et al. Transcutaneous vaccination using a hydrogel patch induces effective
TLR ligands and antigen need to be co-encapsulated into the same immune responses to tetanus and diphtheria toxoid in hairless rat. J
biodegradable microsphere for the generation of potent cytotoxic T Control Release 2010;149:15-20.
lymphocyte responses. Vaccine 2008;26:1626-37. 96. Glenn GM, Taylor DN, Li X, Frankel S, Montemarano A, Alving CR.
75. Shaker DS, Sloat BR, Le UM, Lohr CV, Yanasarn N, Fischer KA, et al. Transcutaneous immunization: a human vaccine delivery strategy using
Immunization by application of DNA vaccine onto a skin area wherein a patch. Nat Med 2000;6:1403-6.
the hair follicles have been induced into anagen-onset stage. Mol Ther 97. Ishii Y, Nakae T, Sakamoto F, Mastuo K, Mastuo K, Quan YS, et al.
2007;15:2037-43. Transcutaneous vaccination system using a hydrogel patch for viral and
76. Vogt A, Combadiere B, Hadam S, Stieler KM, Lademann J, Schaefer bacterial infections. Vaccine 2013;131:113-20.
H, et al. 40 nm, but not 750 or 1,500 nm, NPs enter epidermal CD1a+ 98. Guo L, Qiu Y, Chen J, Zhang S, Xu B, Gao Y. Effective transcutaneous
cells after transcutaneous application on human skin. J Invest Dermatol immunization against hepatitis B virus by a combined approach of
2006;126:1316-22. hydrogel patch formulation and microneedle arrays. Biomed Micro-
77. Hansen S, Lehr CM. NPs for transcutaneous vaccination. Micro devices 2013;15:1077-85.
Biotechnol 2012;5:156-67. 99. Hiraishi Y, Nakagawa T, Quan YS, Kamiyama F, Hirobe S, Okada N, et al.
78. Luengo J, Weiss B, Schneider M, Ehlers A, Stracke F, Konig K, et al. Performance and characteristics evaluation of a sodium hyaluronate-based
Influence of nanoencapsulation on human skin transport of flufenamic microneedle patch for a transcutaneous drug delivery system. Int J Pharm
acid. Skin Pharmacol Physiol 2006;19:190-7. 2013;441:570-9.
79. Stracke F, Weiss B, Lehr CM, Konig K, Schaefer UF, Schneider M. 100. Discher BM, Won YY, Ege DS, Lee JC, Bates FS, Discher DE, et al.
Multiphoton microscopy for the investigation of dermal penetration of Polymersomes: tough vesicles made from diblock copolymers. Science
nanoparticle-borne drugs. J Invest Dermatol 2006;126:2224-33. 1999;284:1143-6.
80. Mattheolabakis G, Lagoumintzis G, Panagi Z, Papadimitriou E, Partidos 101. Bellomo EG, Wyrsta MD, Pakstis L, Pochan DJ, Deming TJ. Stimuli
CD, Avgoustakis K. Transcutaneous delivery of a nanoencapsulated responsive polypeptide vesicles by conformation specific assembly.
antigen: induction of immune responses. Int J Pharm 2010;385:187-93. Nat Mater 2004;3:244-8.
81. Lee PW, Peng SF, Su CJ, Mi FL, Chen HL, Wei MC, et al. The use of 102. Christian DA, Cai S, Bowen DM, Kim Y, Pajerowski D, Dishe DE.
biodegradable polymeric NPs in combination with a low-pressure gene Polymerosome carriers: from self assembly to siRNA and protein
gun for transdermal DNA delivery. Biomaterials 2008;29:742-51. therapeutics. Eur J Pharm Biopharm 2009;71:463-74.
82. Taveira SF, Nomizo A, Lopez RF. Effect of the iontophoresis of a 103. Christian NA, Milone MC, Ranka SS, Li G, Frail PR, Davis KP, et al.
chitosan gel on doxorubicin skin penetration and cytotoxicity. Tat-functionalized near-infrared emissive polymersomes for dendritic
J Control Release 2009;134:35-40. cell labeling. Bioconjug Chem 2007;18:31-40.
M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081 1079

104. Cevc G, Blume G. Lipid vesicles penetrate into intact skin owing to the sustained release vaccine delivery systems. J Liposome Res
transdermal osmotic gradients and hydration force. Biochim Biophys 2012;22:193-204.
Acta 1992;1104:226-32. 127. Bender J, Simonsson C, Smedh M, Engstrom S, Ericson MB. Lipid
105. Benson HA. Transfersomes for transdermal drug delivery. Expert Opin cubic phases in topical drug delivery: visualization of skin distribution
Drug Deliv 2006;3:727-37. using two-photon microscopy. J Control Release 2008;129:163-9.
106. Cevc G. Transfersomes, liposomes and other lipid suspensions on the 128. Kisak E, Coldren B, Zasadzinski JA. Nanocompartments enclosing
skin: permeation enhancement, vesicle penetration, and transdermal vesicles, colloids and macromolecules via interdigitated lipid bilayers.
drug delivery. Crit Rev Ther Drug Carrier Syst 1996;13:257-388. Langmuir 2002;18:284-8.
107. Li N, Peng LH, Chen X, Nakagawa S, Gao JQ. Effective 129. Evans CA, Zasadzinski JA. Encapsulating vesicles and colloids from
transcutaneous immunization by antigen-loaded flexible liposome in cochleate cylinders. Langmuir 2003;19:3109-13.
vivo. Int J Nanomed 2011;6:3241-50. 130. Mishra V, Mahor S, Rawat A, Dubey P, Gupta PN, Singh P, et al.
108. Paul A, Cevc G, Bachhawat BK. Transdermal immunisation with an Development of novel fusogenic vesosomes for transcutaneous
integral membrane component, gap junction protein, by means of immunization. Vaccine 2006;24:5559-70.
ultradeformable drug carriers, transfersomes. Vaccine 1998;16:188-95. 131. Puyal C, Maurin L, Miquel G, Bienvenue A, Philippot J. Design of a
109. Ainbinder D, Touitou E. Testosterone ethosomes for enhanced short membrane-destabilizing peptide covalently bound to liposomes.
transdermal delivery. Drug Deliv 2005;12:297-303. Biochim Biophys Acta 1994;1195:259-66.
110. Fang YP, Tsai YH, Wu PC, Huang YB. Comparison of 5- 132. Delouise LA. Applications of nanotechnology in dermatology. J Invest
aminolevulinic acid-encapsulated liposome versus ethosome for skin Dermatol 2012;132:964-75.
delivery for photodynamic therapy. Int J Pharm 2008;356:144-52. 133. Labouta HI, Schneider M. Tailor-made biofunctionalized NPs using
111. Satyam G, Shivani S, Garima G. Ethosomes: a novel tool for drug layer-by-layer technology. Int J Pharm 2010;395:236-42.
delivery through the skin. J Pharm Res 2011;3:688-91. 134. Xu ZP. Inorganic NPs as carriers for efficient cellular delivery. Chem
112. Rattanapak T, Young K, Rades T, Hook S. Comparative study of Eng Sci 2006;61:1027-40.
liposomes, transfersomes, ethosomes and cubosomes for transcutane- 135. Tiwari PM, Vig K, Dennis VA, Singh SR. Functionalized gold NPs and
ous immunisation: characterisation and in vitro skin penetration. J their biomedical applications. Nanomaterials 2011;1:31-63.
Pharm Pharmacol 2012;64:1509-60. 136. Huang Y, Yu F, Park YS, Wang J, Shin MC, Chung HS, et al. Co-
113. Mishra D, Mishra PK, Dabadghao S, Dubey V, Nahar M, Jain NK. administration of protein drugs with gold NPs to enable percutaneous
Comparative evaluation of hepatitis B surface antigen–loaded elastic delivery. Biomaterials 2010;31:9086-91.
liposomes and ethosomes for human dendritic cell uptake and immune 137. Glenn GM, Rao M, Matyas GR, Alving CR. Skin immunization made
response. Nanomedicine 2010;6:110-8. possible by cholera toxin. Nature 1998;391:851-2.
114. Sjolander S, Drane D, Davis R, Beezum L, Pearse M, Cox J. Intranasal 138. Lipka J, Semmler-Behnke M, Sperling RA, Wenk A, Takenaka S,
immunization with influenza-ISCOM induce strong mucosal as well as Schleh C, et al. Biodistribution of PEG-modified gold NPs following
systemic antibody and cytotoxic T-lymphocyte responses. Vaccine intratracheal instillation and intravenous injection. Biomaterials
2001;19:4072-80. 2010;31:6574-81.
115. Sjolander S, Drane D, Davis R, Beezum L, Pearse M, Cox J. Immune 139. Cho WS, Cho M, Jeong J, Choi M, Han BS, Shin HS, et al. Size-
responses to ISCOM formulations in animals and primate models. dependent tissue kinetics of PEG-coated gold NPs. Toxicol Appl
Vaccine 2000;19:2661-5. Pharmacol 2010;245:116-23.
116. Watson DL, Lovgren K, Watson NA, Fossum C, Morein B, Hoglund S. 140. Bastis NG, Sanchez-Tillo E, Pujals S, Farrera C, Kogan MJ, Giralt E, et
Inflammatory response and antigen localization following immuniza- al. Peptides conjugated to gold NPs induce macrophage activation. Mol
tion with influenza virus ISCOMs. Inflammation 1989;13:641-9. Immunol 2009;46:743-8.
117. Watson DL, Lovgren K, Watson NA, Fossum C, Morein B. Interaction 141. Surujpaul PP, Gutierrez-Wing C, Ocampo-García B, Ramirez Fde M,
between immune stimulating complexes (ISCOMs) and peritoneal Arteaga de Murphy C, Pedraza-Lopez M, et al. Gold NPs conjugated to
mononuclear leucocytes. Microbial Immunol 1992;36:199-203. [Tyr3] Octreotide peptide. Biophys Chem 2008;138:83-90.
118. Maloy KJ, Donachie AM, Mowat AM. Induction of Th1 and Th2 142. Sharma A, Matharu Z, Sumana G, Solanki PR, Kim CG, Malhotra BD.
CD4 + T cell responses by oral or parentral immunization with Antibody immobilized cysteamine functionalized-gold NPs for afla-
ISCOMs. Eur J Immunol 1995;25:2835-41. toxin detection. Thin Solid Films 2010;519:1213-8.
119. Pandey RS, Dixit VK. Evaluation of ISCOMs for immunization against 143. Wangoo N, Bhasin KK, Mehta SK, Suri CR. Synthesis and capping of
hepatitis B. Curr Pharm Biotechnol 2009;10:709-16. water-dispersed gold NPs by an amino acid: bioconjugation and
120. Pandey RS, Babbar AK, Kaul A, Mishra AK, Dixit VK. Evaluation of binding studies. J Colloid Interface Sci 2008;323:247-54.
ISCOM matrices clearance from rabbit nasal cavity by gamma 144. Javier DJ, Castellanos-Gonzalez A, Weigum SE, White AC, Richards-
scintigraphy. Int J Pharm 2010;398:231-6. Kortum R. Oligonucleotide-gold nanoparticle networks for detection of
121. Pandey RS, Dixit VK. Evaluation of ISCOM vaccines for mucosal Cryptosporidium parvum heat shock protein 70 mRNA. J Clin
immunization against hepatitis B. J Drug Target 2010;18:282-91. Microbiol 2009;47:4060-6.
122. Madsen HB, Ifversen P, Madsen F, Brodin B, Hausser I, Nielsen HM. 145. Lee SH, Bae KH, Kim SH, Lee KR, Park TG. Amine functionalized
In vitro cutaneous application of ISCOMs on human skin enhances gold NPs as non cytotoxic and efficient intracellular siRNA delivery
delivery of hydrophobic model compounds through the stratum carriers. Int J Pharm 2008;364:94-101.
corneum. AAPS J 2009;11:728-39. 146. Karande P, Jain A, Mitragotri S. Discovery of transdermal penetration
123. Pan X, Han K, Peng X, Yang Z, Qin L, Zhu C, et al. Nanostructured enhancers by high through put screening. Nat Biotechnol 2004;22:192-7.
cubosomes as advanced drug delivery system. Curr Pharm Des 147. Karande P, Jain A, Ergun K, Kispersky V, Mitragotri S. Design
2013;19:6290-7. principles of chemical penetration enhancers for transdermal drug
124. Duttagupta AS, Chaudhary HM, Jadhav KR, Chilajwar SV, Kadam VJ. delivery. Proc Natl Acad Sci 2005;102:4688-93.
Cubosomes: innovative nanostructures for drug delivery. Curr Drug 148. Mollgaard B. Synergistic effects in percutaneous enhancement. In:
Deliv 2015 [Epub ahead of print]. Walters K, Hadgraft J, editors. Pharmaceutical skin penetration
125. Boyd BJ. Characterization of drug release from cubosomes using the enhancement. New York, Hong Kong, Basel: Marcell Dekker; 1993.
pressure ultrafiltration method. Int J Pharm 2003;260:239-47. p. 229-42.
126. Gordon S, Young K, Wilson R, Rizwan S, Kemp R, Rades T, et al. 149. Williams AC, Barry BW. Penetration enhancers. Adv Drug Deliv Rev
Chitosan hydrogels containing liposomes and cubosomes as particulate 2004;56:603-18.
1080 M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081

150. Baca-Estrada ME, Foldvari M, Ewen C, Badea I, Babiuk LA. Effects of 171. Koutsonanos DG, del Pilar Martin M, Zarnitsyn VG, Sullivan SP,
IL-12 on immune responses induced by transcutaneous immunization Compans RW, Prausnitz MR, et al. Transdermal influenza immuniza-
with antigens formulated in a novel lipid-based biphasic delivery tion with vaccine-coated microneedle arrays. PLoS One 2009;4:e4773.
system. Vaccine 2000;18:1847-54. 172. Zhu Q, Zarnitsyn VG, Ye L, Wen Z, Gao Y, Pan L, et al. Immunization
151. Foldvari M, Kumar P, King M, Batta R, Michel D, Badea I, et al. Gene by vaccine coated microneedle arrays protects against lethal influenza
delivery into human skin in vitro using biphasic lipid vesicles. Curr virus challenge. Proc Natl Acad Sci 2009;106:7968-73.
Drug Deliv 2006;3:89-93. 173. Inoue J, Aramaki Y. Toll-like receptor-9 expression induced by tape-
152. Babiuk S, Baca-Estrada ME, Pontarollo R, Foldvari M. Topical stripping triggers on effective immune response with CpG-oligodeox-
delivery of plasmid DNA using biphasic lipid vesicles (Biphasix). J ynucleotides. Vaccine 2007;25:1007-13.
Pharm Pharmacol 2002;54:1609-14. 174. Huang CM, Wang CC, Kawai M, Barnes S, Elmets CA. Surfactant
153. Heuschkel S, Goebel A, Neubert RH. Microemulsions—modern sodium lauryl sulfate enhances skin vaccination: molecular character-
colloidal carrier for dermal and transdermal drug delivery. J Pharm ization via a novel technique using ultrafiltration capillaries and mass
Sci 2008;97:603-31. spectrometric proteomics. Mol Cell Proteomics 2006;5:523-32.
154. Cui Z, Fountain W, Clark M, Jay M, Mumper RJ. Novel ethanol-in- 175. Huang HN, Li TL, Chan YL, Chen CL, Wu CJ. Transdermal
fluorocarbon microemulsions for topical genetic immunization. Pharm immunization with low-pressure-gene-gun mediated chitosan-based
Res 2003;20:16-23. DNA vaccines against Japanese encephalitis virus. Biomaterials
155. Naoui W, Bolzinger MA, Fenet B, Pelletier J, Valour JP, Kalfat R, et al. 2009;30:6017-25.
Microemulsion microstructure influences the skin delivery of an 176. Prausnitz MR, Bose VG, Langer R, Weaver JC. Electroporation of
hydrophilic drug. Pharm Res 2011;28:1683-95. mammalian skin: a mechanism to enhance transdermal drug delivery.
156. Cui Z, Sloat BR. Topical immunization onto mouse skin using a Proc Natl Acad Sci 1993;90:10504-8.
microemulsion incorporated with an anthrax protective antigen protein- 177. Dahlan A, Alpar HO, Stickings P, Sesardic D, Murdan S. Transcuta-
encoding plasmid. Int J Pharm 2006;317:187-91. neous immunization assisted by low-frequency ultrasound. Int J Pharm
157. Bielinska AU, Janczak KW, Landers JJ, Markovitz DM, Montefiori 2009;368:123-8.
DC, Baker Jr JR. Nasal immunization with a recombinant HIV gp120 178. Tezel A, Paliwal S, Shen Z, Mitragotri S. Low-frequency ultrasound as
and nanoemulsion adjuvant produces Th1 polarized responses and a transcutaneous immunization adjuvant. Vaccine 2005;23:3800-7.
neutralizing antibodies to primary HIV type 1 isolates. AIDS Res Hum 179. Ostberg JR, Gellin C, Patel R, Repasky EA. Regulatory potential of
Retroviruses 2008;24:271-81. fever range whole body hyperthermia on Langerhans cells and
158. Makidon PE, Bielinska AU, Nigavekar SS, Janczak KW, Knowlton J, lymphocytes in an antigen-dependent cellular immune response. J
Scott AJ, et al. Pre-clinical evaluation of a novel nanoemulsion-based Immunol 2001;167:2666-70.
hepatitis B mucosal vaccine. PLoS One 2008;3:e2954. 180. Upadhyay P. Enhanced transdermal-immunization with diptheria-
159. Rutvij JP, Gunjan JP, Bharadia PD, Pandya VM, Modi DA. toxoid using local hyperthermia. Vaccine 2006;24:5593-8.
Nanoemulsion: An advanced concept of dosage form. Int J Pharm 181. Frech SA, Dupont HL, Bourgeois AL, McKenzie R, Belkind-Gerson J,
Cosmetol 2011;1:122-33. Figueroa JF, et al. Use of a patch containing heat-labile toxin from
160. Su X, Kim BS, Kim SR, Hammond PT, Irvine DJ. Layer-by-layer- Escherichia coli against travellers' diarrhoea: a phase II, randomised,
assembled multilayer films for transcutaneous drug and vaccine double-blind, placebo-controlled field trial. Lancet 2008;371:2019-25.
delivery. ACS Nano 2009;3:3719-29. 182. Eypper EH, Johnson PV, Purro EI, Hohmann EL. Transcutaneous
161. Tahara Y, Namatsu K, Kamiya N, Hagimori M, Kamiya S, Arakawa M, immunization of healthy volunteers with an attenuated Listeria
et al. Transcutaneous immunization by a solid-in-oil nanodispersion. monocytogenes vaccine strain and cholera toxin adjuvant. Vaccine
Chem Commun (Camb) 2010;6:9200-2. 2013;31:3201-57.
162. Knockenhauer KE, Sawicka KM, Roemer EJ, Simon SR. Protective 183. U.S. Army Medical Research and Materiel Command. Safety study of
antigen composite nanofibers as a transdermal anthrax vaccine. 30th recombinant vaccine to prevent ETEC diarrhea. ClinicalTrials.gov.
Annual International Conference Proceeding in IEEE Eng Med Biol Bethesda (MD): National Library of Medicine (US); 2000. [Internet]
Soc 2008; 2008. p. 1040-3. [Cited 2015 Sep 9]. Available from: http://clinicaltrials.gov/show/
163. Dubey V, Mishra D, Nahar M, Jain NK. Vesicles as tools for the NCT01382095 NLM Identifier: NCT01382095.
modulation of skin permeability. Expert Opin Drug Deliv 2007;4:579-93. 184. U.S. Army Medical Research and Materiel Command. Safety study of
164. Duplessis J, Egbaria K, Ramachandran C, Weiner N. Topical delivery chimeric vaccine to prevent ETEC diarrhea. ClinicalTrials.gov.
of liposomally encapsulated gamma interferon. Antiviral Res Bethesda (MD): National Library of Medicine (US); 2000. [Internet,
1992;18:259-65. [Cited 2015 Sep 9]. Available from: http://clinicaltrials.gov/show/
165. Bouwstra JA, Honeywell-Nguyen PL. Skin structure and mode of NCT01644565 NLM Identifier: NCT01644565].
action of vesicles. Adv Drug Deliv Rev 2002;54:S41-55. 185. Intercell USA Inc. Traveler's diarrhea (TD) automated process. Clini-
166. Choi MJ, Maibach HI. Liposomes and niosomes as topical drug calTrials.gov. Bethesda (MD): National Library of Medicine (US); 2000.
delivery systems. Skin Pharmacol Physiol 2005;18:209-19. [Internet, [Cited 2015 Sep 9]. Available from: http://clinicaltrials.gov/
167. Honeywell-Nguyen PL, Bouwstra JA. Vesicles as a tool for transdermal show/NCT00751777 NLM Identifier: NCT00751777].
and dermal delivery. Drug Discov Today Technol 2005;2:67-74. 186. Intercell USA Inc. LT vaccine patch self-administration study. Clini-
168. Hofland HE, Bouwstra JA, Ponec M, Bodde HE, Spies F, Coos calTrials.gov. Bethesda (MD): National Library of Medicine (US);
Verhoef J, et al. Interactions of nonionic surfactant vesicles with 2000. [Cited 2015 Sep 9]. Available from: http://clinicaltrials.gov/
cultured keratinocytes and human skin in vitro—a survey of show/NCT00565461 NLM Identifier: NCT00565461. [Internet].
toxicological aspects and ultrastructural changes in stratum corneum. 187. Intercell USA Inc. Gender-stratified safety and immunogenicity study.
J Control Release 1991;16:155-67. ClinicalTrials.gov. Bethesda (MD): National Library of Medicine (US);
169. Cevc G, Schatzlein A, Richardsen H. Ultradeformable lipid vesicles 2000. [Internet, [Cited 2015 Sep 9]. Available from: http://clinicaltrials.
can penetrate the skin and other semi-permeable barriers unfragmented. gov/show/NCT01067781 NLM Identifier: NCT01067781].
Evidence from double label CLSM experiments and direct size 188. University of Zurich. Study of allergen immunotherapy in grass pollen
measurements. Biochim Biophys Acta Biomembr 2002;1564:21-30. allergic subjects with epicutaneous allergen (ZU-SkinSIT-003). Clini-
170. Alarcon JB, Hartley AW, Harvey NG, Mikszta JA. Preclinical calTrials.gov. Bethesda (MD): National Library of Medicine (US); 2000.
evaluation of microneedle technology for intradermal delivery of [Internet, [Cited 2015 Sep 9]. Available from: http://clinicaltrials.gov/
influenza vaccines. Clin Vaccine Immunol 2007;14:375-81. show/NCT00777374 NLM Identifier: NCT00777374].
M. Kaurav et al / Nanomedicine: Nanotechnology, Biology, and Medicine 12 (2016) 1063–1081 1081

189. SIDA Objectif Recherche Vaccins. Safety study comparing a vaccine patients on ART vs Placebo (CUTHIVTHER001). ClinicalTrials.gov.
transcutaneous administration to the intramuscular route (MANON-05). Bethesda (MD): National Library of Medicine (US); 2000. [Internet,
ClinicalTrials.gov. Bethesda (MD): National Library of Medicine (US); Cited 2015 Sep 9. Available from: http://clinicaltrials.gov/show/
2000. [Internet, [Cited 2015 Sep 9]. Available from: http://clinicaltrials. NCT02457689 NLM Identifier: NCT02457689].
gov/show/NCT00261001 NLM Identifier: NCT00261001]. 193. Mayo Clinic, National Cancer Institute. Vaccine therapy and
190. Imperial College London. Immunogenicity of Three HIV GTU® Multi resiquimod in treating patients with stage II, stage III or stage IV
HIV DNA Immunizations Administered Via Intramuscular, Intradermal melanoma that has been completely removed by surgery. Clinical-
and Transcutaneous routes (CUT‫٭‬HIVAC001). ClinicalTrials.gov. Trials.gov. Bethesda (MD): National Library of Medicine (US); 2000.
Bethesda (MD): National Library of Medicine (US); 2000. [Internet, [Internet, [Cited 2015 Sep 9]. Available from: http://clinicaltrials.gov/
[Cited 2015 Sep 9]. Available from: http://clinicaltrials.gov/show/ show/NCT00470379 NLM Identifier: NCT00470379].
NCT02075983 NLM Identifier: NCT02075983]. 194. Slifka MK, Leung Donald YM, Hammarlund E, Raue P, Simpson EL,
191. Combadiere B, Vogt A, Mahe B, Costagliola D, Hadam S, Bonduelle Tofte S. Transcutaneous yellow fever vaccination of subjects with or
O, et al. Preferential amplification of CD8 effector-T cells after without atopic dermatitis. J Allergy Clin Immunol 2014;133:439-47.
transcutaneous application of an inactivated influenza vaccine: a 195. Etchart N, Hennino A, Friede M, Dahel K, Dupouy M, Goujon-Henry
randomized phase I trial. PLoS One 2010;5e10818. C, et al. Safety and efficacy of transcutaneous vaccination using a patch
192. Imperial College London, European Commission. The safety immu- with the live-attenuated measles vaccine in humans. Vaccine
nogenicity of the DNA-GTU Vaccine Administered to HIV-infected 2007;25:6891-9.

View publication stats

S-ar putea să vă placă și