throbber
Page 1 of 12
`
`YEDA EXHIBIT NO. 2039
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`3198
`
`P.H. Lambert, P.E. Laurent / Vaccine 26 (2008) 3197–3208
`
`1. History of intradermal vaccination
`
`The discovery of the principles of vaccination is often described
`as one of the most important developments in public health.
`The practice of inoculating small amounts of material from sick
`patients, such as powdered smallpox scabs or pus, into the nose
`or skin of healthy individuals to prevent disease was widespread
`across parts of Africa, Asia and the Ottoman empire, before inoc-
`ulation into the skin – variolation – was introduced to Europe in
`1721. Inoculated patients would generally develop a milder form of
`the disease than that occurring naturally. However, the risk of death
`from smallpox remained. It was not until 1796 that the first vacci-
`nation was carried out as practiced today by Edward Jenner in the
`United Kingdom based on his observation that milkmaids who had
`contracted cowpox through contact with cowpox pustules were not
`getting smallpox. Initially Jenner’s findings were not well received
`and it took 44 years for variolation to be forbidden by an Act of Par-
`liament and a further 13 years for vaccination against smallpox to
`be made compulsory in Britain in 1853 [1].
`The next important development was made by the French
`physician Charles Mantoux in 1910 when he published his clinical
`research on the intradermal injection of tuberculin as a diagnostic
`skin test for tuberculosis disease [2]. Not only was this technique
`used for tuberculosis diagnosis, but it formed the basis for intra-
`dermal (ID) injection of vaccines, a technique still used today for
`vaccines such as rabies and BCG [3,4].
`In 1967 the WHO launched a global programme to eradicate
`smallpox which, 150 years after Jenner’s discovery, was still affect-
`ing 10–15 million people each year. Eradication of the disease was
`finally confirmed by the World Health Assembly in 1980 [5] A major
`contribution to this achievement was the development of the bifur-
`cated needle by Benjamin A. Rubin. This needle was specifically
`designed to ensure the delivery of about 2 ␮l, but sufficient, quan-
`tity of this very potent vaccine into the dermis. It helped healthcare
`workers to correctly deliver vaccine to the most efficient site for
`immunization against smallpox. Vaccination was done by dipping
`the bifurcated needle into the vial of vaccine to pick up a minute
`drop of vaccine solution between the needle’s two prongs, then by
`jabbing the skin – typically in the deltoid region – several times
`with a brisk movement perpendicularly to the skin surface [6].
`The first renewed interest in intradermal immunization using
`a needle and syringe injection system in controlled clinical trials
`was reported by Tuft in 1930 [7]. This study reported an equiv-
`alent immune response and an improved adverse event profile
`with a smaller dose of typhoid vaccine when injected intrader-
`mally relative to subcutaneous injection [8]. Subsequently to these
`reports several studies aiming to evaluate the efficiency and util-
`ity of intradermal delivery route such as vaccine dose reduction
`were conducted using different commercially available vaccines
`including influenza [9–11], measles [12,13], cholera [14], rabies
`[15,16], hepatitis B [17–20], polio virus [21–24] aiming to evalu-
`ate the optimal route of immunization for preventive vaccination.
`In spite of the large number of published clinical trials compar-
`ing post-immunization humoral immune responses, the evaluation
`of the benefit and utility of intradermal delivery suffers from the
`absence of a consistent clinical design and standardized investi-
`gational method permitting an efficient side-by-side comparison
`and meta-analysis. The vaccine antigen concentration/immune
`response curve has rarely been thoroughly evaluated to detect and
`characterize the minimal, maximal and optimal antigen concen-
`trations in various population segments which correspond to the
`clinical indication of investigated vaccine. Nevertheless, vaccines
`can be generally categorized into three groups: (i) those for which
`intradermal delivery induces better responses than by intramus-
`cular or subcutaneous injection; (ii) vaccines for which conflicting
`
`results has been observed in separate clinical trials; and (iii) vaccine
`remaining to be investigated such as combo vaccines, meningococ-
`cal. Potential benefits of the intradermal delivery route as measured
`by post-immunization immune response depend upon the type
`of vaccine. For example, it is well documented that the immune
`response after intradermal administration of one-tenth of an intra-
`muscular dose is equivalent to the full dose given intramuscularly
`for rabies and hepatitis B vaccines, but not for trivalent influenza
`vaccine [25–29]. One confounding factors leading to mixed clini-
`cal study results with trivalent influenza vaccine is the priming by
`previous natural infection; primed adult subjects produce equiv-
`alent immune response with reduced dose of antigen delivered
`by intramuscular as well as intradermal delivery routes [29,30]. In
`contrast, intradermal influenza vaccination in elderly subjects (15
`and 21 ␮g of haemagluttinin/strain/0.1 ml dose) induced a humoral
`immune response superior to the IM control against all three strains
`[31]. Clinical studies in subjects with chronic medical conditions
`such as kidney failure, with or without haemodialysis, suggest
`that intradermal delivery of hepatitis B vaccine induces a better
`immune response than intramuscular injection [27,28,32]. Meta-
`analysis of clinical trials evaluating rabies vaccine prepared on
`diploid cells indicated that the persistence of specific humoral anti-
`bodies is at least equivalent to that observed with intramuscular
`delivery; the same results are observed with hepatitis B vaccine
`[25,33]. The local skin reactivity usually observed at the injection
`site after intradermal vaccine inoculation reflects the physiological
`local inflammatory response due to immune response induction
`and is characterized by spontaneously reversible redness at the
`injection site for a maximum period of 2 days without local seque-
`lae. Systemic adverse event profiles are equivalent whatever the
`delivery route.
`
`2. Current situation and future needs of innovative vaccine
`delivery systems
`
`An ideal vaccine is safe, cost-effective, and efficient after a sin-
`gle dose [34]. The way in which a vaccine is delivered can have
`considerable bearing on these factors through its influence on the
`efficiency of the procedure, the dose required, compliance, and
`safety. For vaccination to succeed holistically in contributing to
`public health, vaccine delivery systems must allow efficient deliv-
`ery without compromising product stability during storage and
`transport and without negatively influencing patient perception.
`To be considered safe, new delivery systems should reduce the risk
`of injury and infection of healthcare workers, and prevent illicit re-
`use. A delivery system combining all these qualities would facilitate
`the vaccination of greater portion of the population.
`Currently licensed vaccines are delivered via one of five
`main administration routes: intramuscular for the majority of
`vaccines including hepatitis A and B, rabies,
`influenza and
`diphtheria–tetanus–pertussis-based combination vaccines; subcu-
`taneous for vaccines such as measles, mumps and rubella, and
`yellow fever; intradermal for BCG and rabies; intranasal for live
`attenuated influenza vaccine, and oral for poliomyelitis, cholera,
`rotavirus and typhoid fever. With the rare exception of jet injectors,
`intramuscular, subcutaneous, and intradermal routes are accessed
`using needles. These techniques, whilst having proven efficacy in
`terms of achieving the required immune response, have some draw-
`backs relating to safety and patient compliance [35,36]. The invasive
`nature of the parenteral injection procedure and the potential for
`inappropriate reuse of equipment exposes patients to the risk of
`transmission of blood borne pathogens. Additionally, the use and
`disposal of equipment is associated with the risk of needle-stick
`injury. The introduction of safer devices engineered to prevent nee-
`dle re-use and reduce the risk of needle stick infections is likely
`
`Page 2 of 12
`
`YEDA EXHIBIT NO. 2039
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`P.H. Lambert, P.E. Laurent / Vaccine 26 (2008) 3197–3208
`
`3199
`
`to lessen these concerns. However, the perceived or real pain and
`trauma sometimes associated with needle-based vaccination can
`be barriers to vaccination uptake, particularly by needle-phobic
`individuals [37,38]. These drawbacks, and the development of new
`types of vaccines, are some of the reasons driving the pharmaceu-
`tical industry and public health organizations to search for new
`delivery methods that are safe, cost-effective and efficient.
`While the majority of vaccines in clinical development are envi-
`sioned as needle and syringe products, a number of research groups
`and vaccine manufacturers are exploring the advantages of new
`parenteral delivery systems as well as of mucosal and transcu-
`taneous delivery [39]. Mucosal delivery is currently only used
`for live attenuated vaccines against poliomyelitis, typhoid fever
`(oral), rotavirus and influenza (nasal) [40,41]. Mucosally adminis-
`tered vaccines have a number of benefits. They eliminate the risk
`of transmission of blood borne diseases and needle stick injury.
`They can potentially be given by personnel with little medical
`training, which provides significant practical and cost benefits, par-
`ticularly in the context of large-scale immunisation programmes
`in the developing world [42]. This route can also, in theory, elicit
`both mucosal and humoral immunity, offering advantages against
`diseases contracted via mucosal surfaces [43]. However, there
`are also a number of drawbacks. The live attenuated viruses in
`oral poliomyelitis vaccine (OPV) can revert to virulence, causing
`vaccine-associated paralytic poliomyelitis (VAPP) in the vaccinated
`child or their close contacts, particularly in the immuno-depressed
`subjects [44]. This has resulted in a shift from the use of OPV
`to the use of injectable poliomyelitis vaccine containing inacti-
`vated virus, especially in countries that have eliminated naturally
`occurring polio [45]. Oral vaccines have to overcome problems
`associated with poor absorption or degradation within the diges-
`tive system that may require the concomitant administration of
`antacids [46]. Finally, to date no mucosal vaccine adjuvant is
`available with the required safety and efficacy [44]. Such safety
`issues were encountered with an intranasal adjuvant-containing
`influenza vaccine that was associated with the occurrence of facial
`palsy [47].
`
`3. Skin physiology and immunology
`
`3.1. Skin anatomy
`
`An increasing understanding of skin physiology means that this
`organ is now recognized as a potentially excellent site for vacci-
`nation. It is easily accessible and has both cellular and humoral
`immune system components. The skin is comprised of three
`primary layers from outside to inside: epidermis, dermis and
`hypodermis (Fig. 1). Vaccine delivery into these layers is known,
`respectively as transdermal, intradermal and subcutaneous vacci-
`nation.
`The epidermis is the outermost layer of the skin and acts as a
`physical barrier, preventing chemicals and micro-organisms from
`entering the body and stopping excess body water loss. This layer
`is generally 50–200 ␮m thick, depending on the body region and
`has four sublayers: the outermost stratum corneum, below which
`is the stratum granulosum, the stratum of Malpighii or spinosum,
`and finally the stratum basale (or germinativium). Keratinocytes
`constitute approximately 90% of the epidermis; the remaining cells
`are melanocytes and Langerhans dendritic cells. While Langerhans
`cells account for only about 1% of cells, they cover nearly 20% of
`the surface area due to their horizontal orientation and long pro-
`trusions [48]. The epidermis does not have its own blood supply;
`cells in lower levels receive nutrients via diffusion from blood cap-
`illaries in the dermis. Cells form within the stratum basale and
`migrate through to the stratum corneum where they are sloughed
`off. During this process, which lasts approximately 30 days, cells
`become keratinised. It is the stratum corneum with its layer of ker-
`atinised cells that is so important in the skin’s role as a physical
`barrier. The stratum corneum is also the greatest barrier to effec-
`tive transdermal vaccine delivery. To be effective, it is critical that
`the vaccine be delivered to the Langerhans cells. This implies that
`a transdermal delivery method must include a system to disrupt,
`either physically or chemically, the stratum corneum, allowing anti-
`gens to pass through this layer and onto the Langerhans cells for
`antigen presentation.
`
`Fig. 1. Skin anatomy. Skin thickness was measured by 20 MHz ultrasound echography in usual body sites for vaccine delivery [55].
`
`Page 3 of 12
`
`YEDA EXHIBIT NO. 2039
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`3200
`
`P.H. Lambert, P.E. Laurent / Vaccine 26 (2008) 3197–3208
`
`The dermis lies beneath the epidermis and is comprised of col-
`lagen, elastin and reticular fibres. It is a tough, flexible and very
`elastic layer between 1.5 and 3 mm thick, arranged into two sub-
`layers: the papillary dermis and the reticular dermis. The papillary
`dermis is the upper and the thinner of the two and consists of
`loosely arranged connective tissue. The reticular dermis consists
`of a network of horizontally running collagen fibres, connective
`tissue, and a very dense network of capillary blood and lymphatic
`vessels in which dermal dendritic cells, monocytes, polymorphonu-
`clear lymphocytes and mast cells circulate. Lymphatic vessels drain
`the dermis to satellite lymph nodes. Fibroblasts are the most abun-
`dant type of cells in the dermis. Endothelial cells forming the wall of
`blood and lymphatic channels play a key role in the inflammatory
`and immune cells as well as fluid movements in dermis. Endothelial
`cell contribute to various physiological effects in the skin including
`vasodilation increased permeability, increased vasomotion, pro-
`duction of cytokines converting adherent leukocytes into mobile
`cells, angiogenesis and trafficking of antigen presenting cells, T and
`B effector cells [49].
`The hypodermis, or subcutaneous tissue, is a layer of loose con-
`nective tissue and elastin located immediately beneath the dermis.
`The arteries and veins that drain the skin dermis issue from the
`vascular plexus located in subcutaneous tissue. When entering the
`skin dermal arteries form a dense network of capillary loops in the
`papillary dermis layer. Numerous lymphatic vessels draining the
`skin dermis pass through the hypodermis before reaching draining
`lymph nodes. The hypodermis is the main tissue for fat storage.
`Anatomical variations of skin according to body site, gender,
`age and ethnic origin are important parameters to consider for
`dermal vaccination. For example, skin thickness – an essential
`parameter for intradermal vaccination – is known to vary signif-
`icantly between different parts of the body [50–54]. In a recent
`study designed specifically to investigate skin thickness at the usual
`areas for intradermal vaccination (deltoid, suprascapular, upper
`abdomen and thigh) in groups of people of different age, sex and
`ethnic origin, skin was found to be on average 1.5 mm thick at the
`thigh and between 1.8 mm and 2.7 mm at the other body sites,
`with no major differences between the different population sub-
`groups considered [55]. Indeed skin thickness was found to vary
`less between people of different body mass index, age, gender and
`ethnic origin than it did between different body sites on people
`with the same demographic characteristics [55]. The average thick-
`ness of the skin appears to remain relatively unchanged in the age
`range of 18–70 years [56]. Skin is thinner in women than in men
`
`by 0.06–0.2 mm, but minimal skin thickness in women is greater
`than 1.5 mm in all cases [55–58]. The absence of a significant effect
`of the ethnic origin on the skin thickness at deltoid, and supras-
`capular body sites has also been reported in studies in US [54]
`and Japan [59]. This consistency in skin thickness across people
`with different demographic profiles represents a major advantage
`over classic intramuscular vaccination as, to correctly perform an
`intramuscular vaccination, it is important to select the appropriate
`needle length based on considerations of the muscle mass of the
`injection site, the amount of subcutaneous fat, and the weight of
`the patient [52,60].
`
`3.2. Skin and immune response
`
`The skin generates both innate (antigen non-specific response
`without immunological memory) and adaptive immune responses
`(antigen specific response with immunological memory), Table 1.
`While the adaptive response is primordial in generating a response
`to vaccination and generally becomes more effective with each
`successive encounter with an antigen [63], innate immune mech-
`anisms also play a key role as they are activated first in response to
`pathogen invasion or contact with foreign antigens. The key group
`of immune cells involved in the skin’s innate immune response is
`dendritic leukocytes: Langerhans cells in the epidermis and dermal
`dendritic cells in the dermis [63–68].
`In 1868 Paul Langerhans, driven by the interest in the anatomy
`of skin nerves, identified a population of dendritically shaped
`cells in the suprabasal region of the epidermis after impregnat-
`ing human skin with gold salt [65]. These cells are known as
`antigen-presenting cells, called Langerhans cells after their discov-
`erer. Although substantial numbers of dendritic leukocytes reside
`and circulate in the skin, only some of them are Langerhans cells,
`the majority being phenotypically different from Langerhans cells
`and generically called dermal dendritic cells [64]. Both Langerhans
`cells and dermal dendritic cells are bone marrow-derived leuko-
`cytes highly specialized in antigen-presenting properties. These
`cells, in association with macrophages recruited from circulating
`blood and infiltrating dermis tissue, are the gatekeepers of the
`immune systems. Compelling evidence exists that Langerhans cells
`and dermal dendritic cells, as members of the family of antigen-
`presenting cells play a pivotal role in the induction of adaptive
`immune response against pathogens and any other antigens and
`haptens which compromise the host homeostasis. The immuno-
`genic potential of antigen-presenting cells from both epidermis and
`
`Table 1
`Innate and adaptive skin immune system
`
`Functional components of skin immune system
`
`Cells of the skin immune system
`
`Innate
`Reactive oxygens
`Ligands Toll receptors
`Heat shock proteins
`Cytokines: IL-1, IL-6, TNF␣
`Chimiokines: CC, CXC
`Adhesion molecules
`Neuropeptides
`Eicosanoids
`Immune tolerance: T regulator, IL-10, TGF␤
`
`Adaptive
`Antigen recognition and presentation
`Cytokines: IL-1, IL-6, TNF␣
`IL-2, IL-12, IL-18, INF␣
`Chimiokines: CC, CXC
`Adhesion molecules
`T and B lymphocyte responses with high affinity effectors
`
`Resident
`
`Recruited
`
`Recirculating
`
`Keratinocytes
`Endothelial cells
`Langerhans cells
`
`Monocytes
`Granulocytes
`
`Natural killer cells
`Dendritic cells
`
`Mast cells
`Macrophages
`T lymphocytes
`Dendritic cells
`
`Mast cells
`Epitheloid cells
`T lymphocytes
`B lymphocytes
`
`Pro-monocytes
`
`T lymphocytes
`
`Page 4 of 12
`
`YEDA EXHIBIT NO. 2039
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`P.H. Lambert, P.E. Laurent / Vaccine 26 (2008) 3197–3208
`
`3201
`
`dermis tissues is regulated by cell surface receptors triggered by lig-
`ands secreted or presented by other somatic cells, or alternatively,
`by microbial products (danger or competence signals) [66]. Dan-
`ger signals are represented for example by DNA rich in CpG repeats
`in bacteria, or other Toll-like receptor ligands [66,67]. Many of the
`receptor structures that sense such signals are essential compo-
`nents of the innate immune system. They are used to recognize
`molecular patterns demarking infectious nonself, as well as normal
`and abnormal self. The response to danger signals leads to tissue
`perturbation as evidenced by increased secretion of GM-CSF, TNF-
`␣, IL-1 by keratinocytes and other skin cells. The antigen-presenting
`cells that pick up the antigen, process it, and re-express part of it as
`peptide/MHC complex on the surface are also profoundly affected
`by danger signals or danger signal-induced cytokine. The alter-
`ations of Langerhans cells and dermal dendritic cells include the
`increased expression of MHC antigens, co-stimulatory molecules,
`and cytokines such as IL-1␤, IL-6, IL-12, as well as the enhanced
`emigration of these cells from the skin to the paracortical area
`of draining lymph nodes. At this site, the skin-derived dendritic
`cells provide the activation stimuli to na¨ıve resting T cells sur-
`rounding them. This occurs in an antigen-specific fashion and
`thus results in the expansion of the respective clone(s) to mature
`into extremely potent immuno-stimulatory cells that controls the
`development of adaptive immunity [68]. Some evidence also exists
`that dermal dendritic cells that have not received such compe-
`tence signals are not stimulatory, but actively down regulate or
`prevent potentially harmful immune responses by tolerizing T cells
`or by inducing T cells with suppressive properties (regulatory T
`cells) [69,70]. Several studies have indicated that protein or pep-
`tide delivery through the epidermis can lead to production of
`specific IgE due to a Th2-regulated response as well as immune
`tolerance status by regulatory T-cells [71–75]. As a consequence,
`in addition to the immuno-surveillance activity, the skin immune
`system secures the homeostasis of the skin integument by prevent-
`ing the development of exaggerated, tissue destructive immune
`responses against per se innocuous moieties such as auto-antigens,
`allergens and haptens. Interestingly, a clinical study in healthy
`adults evaluating epidermal delivery of live-attenuated measles
`vaccine through disrupted stratum corneum relative to intra-
`muscular route strongly suggests that resident antigen-presenting
`cells in epidermis were unable to boost the antibody response
`[76].
`
`3.3. Skin immune response and sun exposure
`
`It has been suggested that sun exposure may affect local
`or systemic immune responses through release of inflammatory
`mediators [80]. The question may be raised whether such effects
`may particularly influence responses to intradermal immunization.
`UV radiation below 290 nm is absorbed by the ozone layer in the
`stratosphere and does not reach the Earth’s surface. The UVB wave-
`lengths range from 280 to 315 nm and from 315 to 400 nm for UVA.
`Solar UV radiation is 95–98% UVA and 25% UVB. The most obvious
`clinical effects of the sun exposure are sunburn and tanning, but
`include more complex biological effects such as DNA photo dam-
`age, immunosuppression and vitamin D synthesis. These biological
`effects are radiation dose-dependent and the amount of UV radia-
`tion penetrating the epidermis and dermis is the critical factor. For
`instance, the stratum corneum of the epidermis is able to dissipate
`90% of UVB radiation, and no more than 10% of UVB reaches the
`dermal-epidermal junction area. In addition, melanin present in
`high concentration in the epidermis acts as UV radiation filter. The
`biological effects of UVB on the skin immune response was actively
`investigated, the main changes being the depletion of Langerhans
`cells, the increased recruitment of macrophages in skin dermis and
`
`the release of pro-inflammatory cytokines such as TNF-␣, IL-10,
`TGF-␤, ␣-MSH and CGRP [77–80].
`
`4. Clinical experience, techniques and devices for
`intradermal vaccination
`
`Considerable clinical research has been conducted to compare
`the intradermal route with other routes of vaccine delivery (Table 2)
`and into new techniques for intradermal delivery to eliminate some
`of the problems associated with the methods currently available.
`This section will describe the available techniques, as well as those
`in clinical research or earlier development.
`
`4.1. Current methods
`
`4.1.1. Mantoux injection technique
`The standard intradermal
`injection technique consists of
`stretching the surface of the skin and inserting the tip of a 27G,
`3/8 in. short bevel needle attached to a plastic 1 ml disposable
`syringe. The needle is inserted bevel upwards, almost parallel to
`the skin surface and vaccine is injected slowly into the uppermost
`layer of the skin [81]. If placed properly, there is considerable resis-
`tance to injection and a raised papule immediately appears which
`can cause pain during injection. The correct placing of the needle-
`tip in the dermis is critical to avoid fluid injection difficulties due
`to inelastic skin or age-related anatomic changes [53,58,61,62,81].
`This technique, introduced by Charles Mantoux over 95 years ago as
`a diagnostic skin test for tuberculosis disease [2] has not been pur-
`sued for the vast majority of vaccines due to its inherent difficulties.
`This technique is associated with a poor consistency of the injected
`volume, due in part to the difficulty of performing it correctly, but
`also to the unavoidable leakage of vaccine from the injection site,
`fluid wastage when filling disposable syringes and when purging
`the needle of air, and the large dead volume of the assembled dis-
`posable needle and syringe [82–84]. In many cases, intradermal
`vaccination according to Mantoux has proved to be comparably
`immunogenic to the comparator even at a reduced dose, due to the
`skin’s ability to generate a strong immune response [3,27,85–91].
`This comparable efficacy at lower doses suggests that intradermal
`injection can have considerable benefits over other injection tech-
`niques when mass vaccination is necessary, as the reduced dose
`means improvement of vaccine availability and of health economic
`ratios if an injection system that is easier to practice becomes avail-
`able.
`
`4.1.2. Bifurcated needle
`While working for Wyeth Laboratories in 1965, Benjamin Rubin
`developed his two-pronged needle for smallpox vaccination by skin
`scarification by grinding the eyelet of a sewing machine needle into
`a fork shape. This was the first example in modern medical history
`of a device specifically designed to deliver vaccine intradermally.
`The small space between the two tines was able to hold about 2 ␮l of
`vaccine solution but only part of this volume that was actually intro-
`duced into the skin and precise control of dose delivery accuracy
`was not possible. The needle was jabbed into the papillary dermis
`skin layer, yielding a spot of blood. Bifurcated needles with features
`for protecting health-care workers against needle-stick injuries are
`commercially available.
`
`4.1.3. Multipuncture
`The percutaneous BCG delivery using single or multipunc-
`ture devices was introduced by Sol Roy Rosenthal in 1939, and
`developed worldwide by the Merieux Institute [92–94]. The mul-
`tipuncture unit is a cylinder-like device with small needles, 1 mm
`length, which should be pressed firmly against the skin, within the
`
`Page 5 of 12
`
`YEDA EXHIBIT NO. 2039
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`3202
`
`P.H. Lambert, P.E. Laurent / Vaccine 26 (2008) 3197–3208
`
`area where the vaccine dose is spread on the skin surface. In con-
`trast with the intradermal technique, the percutaneous method
`does not allow a precise estimation of the dose injected in the
`epidermis or dermis skin layers. As a consequence, the num-
`ber of live attenuated bacilli given per delivery is highly variable
`[95–97].
`
`4.1.4. Needle-free jet injection
`Needle-free jet injection uses a gas to force a liquid or powder
`vaccine through the skin, delivering it intradermally, intramus-
`cularly or subcutaneously [98,99]. These devices can either be
`multidose or monodose. The latter are based either on entirely dis-
`posable devices or on a re-usable system with single dose cartridges
`[99]. This technology has been available since the 1940s and was
`used for several mass vaccination programmes [98–101]. However,
`when an outbreak of hepatitis B linked to the use of multidose
`jet injectors revealed the risk of contamination by the aspiration
`of small amounts of blood into the nozzle during the vaccination
`procedure, their use was discontinued by the WHO and health
`authorities around the world [101]. Recent research has focused
`on improved devices with single-use nozzles to remedy this prob-
`lem [102] and on developing standardised single-dose cartridges
`that could potentially make monodose injectors financially viable
`for mass vaccination [99].
`Studies have shown that vaccination via jet injection can elicit
`an immune response comparable to or better than other delivery
`methods. A comparison of jet injection with the standard needle-
`based techniques for five vaccines found that for four of them,
`jet injection had equivalent, if not improved efficacy [97,98,104].
`Successful intramuscular or subcutaneous jet injection has been
`demonstrated with a wide range of vaccine technologies includ-
`ing DNA coated nanoparticles [105], naked DNA [106], inactivated
`virus, polysaccharide–protein conjugates, toxoids, and whole cell
`vaccines [103]. In one study with tetanus toxoid vaccine, jet injec-
`
`tion was found to be less immunogenic than the standard needle
`and syringe technique in subjects younger than 40 years, although
`the techniques gave equivalent results in those aged over 40 [102].
`In this study the jet injector delivered the vaccine into the deep
`subcutaneous layer.
`In contrast to needle-based vaccination, jet injection results in a
`wide distribution of vaccine in the dermis, hypodermis and muscle,
`depending on jet injector settings and individual body character-
`istics. This distribution effect means that jet injection may face
`some problems when used to specifically deliver a precise dose
`of vaccine into the dermis, but is thought to lead to the increased
`immune responses observed [102,108]. It is also possible that the
`larger immune responses are a result of increased inflammation
`with jet injectors, leading to the recruitment of more immune cells
`to the injection site [107,108]. Indeed, while it has been suggested
`that jet injection results in less tissue damage than with needles
`as the injected liquid follows the path of least resistance [109] a
`number of studies refute this finding that jet injection (whether
`intradermal, intramuscular or subcutaneous) causes more adverse
`events including swelling, erythema, induration, haematoma and
`pain [98,103,110,111]. In a comparison of DNA vaccine delivered
`either by intramuscular injection or by intradermal jet injection, or
`a combination of the two, jet injection caused approximately twice
`as many adverse events. Despite this, the majority of subjects in this
`study stated that they preferred the jet injection to needle injection
`at the time of injection [106].
`Jet injection with single use nozzles or single dose cartridges
`removes the risk of needle stick injuries after vaccine delivery and
`associated risk of blood borne transmission of diseases through
`contamination. In addition, the nature of jet injection is such that a
`large number of vaccines (in excess of 600 per hour) can potentially
`be treated in a short period of time [102]. This makes jet injectors an
`attractive technology for mass parenteral vaccination programmes.
`It has indeed been used in a number of mass campaigns for small-
`
`Table 2
`New technologies targeting vaccine delivery into the skin
`
`Technology
`
`Prefilled microinjection
`system
`
`Non-prefilled microinjection
`needle
`
`Company
`
`BD Medical Pharmaceutical Systems/Sanofi Pasteur
`
`BD Medical Pharmaceutical Systems/Oncovax
`
`Nanopass, Micro-Pyremidal Needle
`Georgia Institute of Technology
`Debiotec
`
`Topical Patch with cholera toxin adjuvant
`
`Iomai Corporation
`
`Topical Patch
`
`Transdermal with electroporation
`
`Solid microneedle array
`
`Vaxin, Inc.
`Ichor Medical Systems, TriGrid
`
`Cyto Pulse: Derma Vax, Easy Vax
`Inovio, MedPulser DNA Delivery System
`
`Genetronics Biomedical Corporation
`Alza Corporation, Macroflux
`
`Biovalve, Micro-Trans
`Epidermal Powder Immunization
`
`Jet injector (Powder)
`
`PowderMed
`
`Jet injector (liquid)
`Skin abrasion
`Skin permeation by low frequency (20 kHz)
`ultrasound associated with topical patch
`Nanoparticle and microparticle
`formulation
`
`Bioject
`BD Technology, Microenhancer Array
`Sonics & Materials, Newtown, CT
`
`Chiron
`Various
`
`Vaccine (development phase)
`
`Trivalent inactivated seasonal
`influenza (clinical phase 3)
`Cancer vaccine (clinical phase 2)
`
`Flu (clinical phase 1)
`Pre-clinical
`Pre-clinical
`
`Trivalent inactivated seasonal
`influenza (clinical phase 2) Travelers’
`diarrhea (clinical phase 2)
`
`Pre-clinical
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket