Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Annals of Medicine

ISSN: 0785-3890 (Print) 1365-2060 (Online) Journal homepage: http://www.tandfonline.com/loi/iann20

Understanding modern-day vaccines: what you


need to know

Volker Vetter, Gülhan Denizer, Leonard R. Friedland, Jyothsna Krishnan &


Marla Shapiro

To cite this article: Volker Vetter, Gülhan Denizer, Leonard R. Friedland, Jyothsna Krishnan &
Marla Shapiro (2017): Understanding modern-day vaccines: what you need to know, Annals of
Medicine, DOI: 10.1080/07853890.2017.1407035

To link to this article: https://doi.org/10.1080/07853890.2017.1407035

View supplementary material

Published online: 27 Nov 2017.

Submit your article to this journal

Article views: 79

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iann20

Download by: [University of Florida] Date: 04 December 2017, At: 02:10


ANNALS OF MEDICINE, 2017
https://doi.org/10.1080/07853890.2017.1407035

REVIEW ARTICLE

Understanding modern-day vaccines: what you need to know


Volker Vettera €lhan Denizerb, Leonard R. Friedlandc
, Gu , Jyothsna Krishnana and Marla Shapirod
a
R&D Department, GSK, Wavre, Belgium; bRegulatory Affairs Department, MSD, Brussels, Belgium; cR&D Department, GSK, Navy Yard,
Philadelphia, PA, USA; dDepartment of Family and Community Medicine, University of Toronto, Toronto, Canada

ABSTRACT ARTICLE HISTORY


Vaccines are considered to be one of the greatest public health achievements of the last cen- Received 4 September 2017
tury. Depending on the biology of the infection, the disease to be prevented, and the targeted Revised 7 November 2017
population, a vaccine may require the induction of different adaptive immune mechanisms to Accepted 14 November 2017
be effective. Understanding the basic concepts of different vaccines is therefore crucial to under-
stand their mode of action, benefits, risks, and their potential real-life impact on protection. This KEYWORDS
review aims to provide healthcare professionals with background information about the main Benefit; concept; design;
vaccine designs and concepts of protection in a simplified way to improve their knowledge and
Downloaded by [University of Florida] at 02:10 04 December 2017

immune response;
understanding, and increase their confidence in the science of vaccination (Supplementary limitation; vaccinology
Material).

KEY MESSAGE
 Different vaccine designs, each with different advantages and limitations, can be applied for
protection against a particular disease.
 Vaccines may contain live-attenuated pathogens, inactivated pathogens, or only parts of
pathogens and may also contain adjuvants to stimulate the immune responses.
 This review explains the mode of action, benefits, risks and real-life impact of vaccines by
highlighting key vaccine concepts.
 An improved knowledge and understanding of the main vaccine designs and concepts of
protection will help support the appropriate use and expectations of vaccines, increase confi-
dence in the science of vaccination, and help reduce vaccine hesitancy.

1. Introduction the risk of acquiring the disease and its potential


complications.
Vaccines are one of the greatest public health achieve-
ments of the last century and are estimated to save
2–3 million lives each year [1]. They have successfully 1.2. How do vaccines work?
eradicated smallpox and have greatly reduced the inci-
Vaccines, like natural infections, act by initiating an
dence of several major diseases such as polio and innate immune response, which in turn activates an
measles [1,2]. Licensed vaccines are now available to antigen-specific adaptive immune response [3]. Innate
prevent over 30 different infectious diseases, several of immunity is the first line of defence against pathogens
which can be combined into single vaccines or admin- that have entered the body. It is established within a
istered at a single vaccination visit [1]. In this review, few hours but is not specific for a particular pathogen
we highlight the different vaccine designs and illus- and has no memory [4]. Adaptive immunity provides a
trate them through various examples to give second line of defence, generally at a later stage of
non-experts a basic understanding of vaccines and infection, characterized by an extraordinarily diverse
concepts of prevention. set of lymphocytes and antibodies able to recognize
and eliminate virtually all known pathogens. Each
pathogen (or vaccine) expresses (or contains) antigens
1.1. What is the aim of vaccination?
that induce cell-mediated immunity by activating
The aim of vaccination is to induce a protective highly specific subsets of T lymphocytes and humoral
immune response to the targeted pathogen without immunity by stimulating B lymphocytes to produce

CONTACT Volker Vetter volker.v.vetter@gsk.com GSK, Building W23-D2, 20 Avenue Fleming, 1300 Wavre, Belgium
Supplemental data for this article can be accessed here.
ß 2017 Informa UK Limited, trading as Taylor & Francis Group
2 V. VETTER ET AL.

specific antibodies [3]. After elimination of the patho- could contribute to vaccine hesitancy among health-
gen, the adaptive immune system generally estab- care providers or patients [9].
lishes immunological memory. This immunological
memory – the basis of long-term protection and the
2. Live attenuated vaccines
goal of vaccination – is characterized by the persist-
ence of antibodies and the generation of memory cells 2.1. What are live attenuated vaccines?
that can rapidly reactivate upon subsequent exposure
Live attenuated vaccines contain pathogens that have
to the same pathogen [3].
been weakened, altered or selected to be less virulent
than their wild-type counterparts. In their altered form,
1.3. What you need to know about vaccine design they cannot cause the actual disease or only mimic
and concepts? the disease in a very mild way.
Live attenuated vaccines are generally produced
Vaccine design has made significant advances in the
from viruses rather than bacteria because viruses con-
last century, evolving from serendipity to a more
tain fewer genes and attenuation can be obtained and
rational design due to advances in understanding
controlled more reliably. The most common method
immunological mechanisms and technology [1].
to obtain live attenuated vaccines is to pass the virus
Depending on the biology of the infection and the
Downloaded by [University of Florida] at 02:10 04 December 2017

through a series of in vitro cell cultures (e.g. in chick


disease to be prevented, a vaccine may require the
embryo cells) [10]. At each “passage”, the selected
induction of different humoral (i.e. antibodies) or cell-
viruses become better at infecting and replicating in
mediated (i.e. T cells) adaptive immune mechanisms to
be effective. Understanding the mode of action of vac- cell cultures but progressively lose their ability to
cines is therefore important to predict their efficacy, infect and replicate in their original human host.
their safety profile, and their expected benefit for the Attenuation can also be achieved by low-temperature
vaccinated individuals and the general population. passages (e.g. 25  C) [10]. This approach selects viruses
Although vaccines are mostly seen as tools for that replicate well in a cold environment but less well
individual protection, vaccines can also protect unvac- at body temperature, therefore decreasing their patho-
cinated populations by reducing the rate of person- genicity in the human host.
to-person transmission and limiting the risk for individ- Live attenuated vaccines act by causing a very lim-
uals to be exposed. This indirect protection, called ited type of infection. As the attenuated pathogen
herd or community protection, requires that a large presents the same antigens as the original pathogen,
portion of the population (75–95% depending on the healthy individuals develop immune responses com-
disease), or a special group that plays a key role in parable to those induced by the natural infection.
transmission of the disease, is vaccinated [5,6]. Herd Consequently, these vaccines induce robust cell-medi-
protection is often essential for the success of vaccin- ated and antibody responses and often confer long-
ation programs, such as for measles [7]. Similarly, vac- term immunity after only one or two doses [11].
cination of pregnant women can also indirectly Another advantage of live viral vaccines, and a risk
protect infants in their first months of life through also, is that they can induce herd immunity through
transfer of maternal antibodies from the mother to the excretion of viral particles (i.e. viral shedding), which
foetus across the placenta [8]. This concept has may indirectly “vaccinate” individuals living in the
been successfully established for tetanus, influenza environment of the vaccine. This phenomenon differs
and pertussis [8]. from the herd protection induced by vaccinating
In contrast to the generally well-known pharmaco- enough individuals to stop transmission of the patho-
logical effects of different drugs, the differences gen but may be also important to achieve a high pro-
between vaccine types are also important but less well tection level.
understood by many healthcare providers. Different Live attenuated vaccines have some limitations.
vaccines targeting the same pathogen can rely on Although rare, clinical disease can occur after vaccin-
very different concepts (Figure 1), each having advan- ation, but vaccine-induced symptoms are typically
tages and limitations (Table 1). Choosing a particular much milder than after natural infection. However,
vaccine therefore can depend on several factors such immunocompromised individuals are at risk of unregu-
as the level of protection, the expected mode of lated pathogen replication that may lead to severe
action, the characteristics of the subject, or the disease infection or death. Therefore, live attenuated vaccines
elimination strategy. Many healthcare providers have are often contraindicated in immunocompromised
not received sufficient education on vaccines, which individuals, and these individuals should only receive
ANNALS OF MEDICINE 3
Downloaded by [University of Florida] at 02:10 04 December 2017

Figure 1. Different types of vaccines. Vaccines can be produced using different processes. Vaccines may contain live attenuated
pathogens (usually viruses), inactivated whole pathogens, toxoids (an inactivated form of the toxin produced by bacteria that
causes the disease), or parts of the pathogens (e.g. natural or recombinant proteins, polysaccharides, conjugated polysaccharide or
virus-like particles).

live attenuated vaccines after a critical benefit–risk All currently-used yellow fever vaccines derive from
assessment considering the type and severity of this attenuated strain [13]. In addition, YF-17D has
immunodeficiency. Similarly, live attenuated vaccines been used to produce vaccines against two closely
are also contraindicated during pregnancy due to the related viruses, Japanese encephalitis (IMOJEV, Sanofi
theoretical risk of foetal infection that may result in Pasteur) and dengue (Dengvaxia, Sanofi Pasteur), by
congenital disease (e.g. rubella or varicella) [12]. replacing the genes encoding the antigenic proteins
Finally, another consideration is that live attenuated by their equivalents [14,15].
vaccines have the potential to revert back to a form Other classical examples of live attenuated vaccines
able to cause the disease, though this is very rare (e.g. produced by serial passage are those against measles,
oral polio vaccine [OPV]). mumps, rubella and varicella, which are usually com-
bined into trivalent (Priorix, GSK; M-M-RVAXPRO, MSD)
or tetravalent (Priorix-Tetra, GSK; ProQuad, MSD) vac-
2.2. Which vaccines are live attenuated? cines [16,17]. The live attenuated varicella vaccine
Many of the first vaccines that were produced con- strain is also used in the herpes zoster (shingles) vac-
sisted of live attenuated vaccines, such as rabies, cine (Zostavax, MSD) but this vaccine relies on a differ-
smallpox, tuberculosis, yellow fever and OPV, some of ent rationale. It contains a high dose of the live
which are still in use. A successful example is the yel- attenuated varicella-zoster virus (>14 times more than
low fever vaccine YF-17D. It was developed in the in the varicella vaccine) to boost immune responses in
1930s by attenuating a yellow fever virus strain by older adults who are already immune to varicella to
more than 200 serial passages through monkeys and prevent viral reactivation and the development of
cultures of mouse and chicken embryonic tissues [13]. shingles [18].
4 V. VETTER ET AL.

Table 1. Benefits and limitations of the different types of vaccines.


Vaccine type Benefits [27,68] Limitations [27,68] Examples
Live attenuated vaccines
Mimic natural infection and immune Contraindicated for immunocomprom- Measles, mumps, rubella, varicella
response ised individuals and pregnant (Priorix Tetra, ProQuad), rotavirus
women (Rotarix, Rotavac), herpes zoster
Elicit both antibodies and cell-medi- Less stable over time, heat labile (Zostavax), influenza (Flumist), oral
ated immunity poliovirus (OPV), yellow fever
Life-long immunity possible after 1 or Possibility to reverse to natural form (Stamaril)
2 doses (e.g. poliovirus)
Inactivated vaccines
Robust technique for production Limited immunogenicity, adjuvants Whole-cell pertussis (Tritanrix),
may be required hepatitis A (Havrix, Vaqta), rabies
High stability Multiple primary and booster doses (Rabipur), tickborne encephalitis
required to obtain long-term (Encepur), Japanese encephalitis
protection (Ixiaro), cholera (Dukoral)
Impact on carriage
No replication of the inactivated
pathogen
Not contraindicated in immunocom-
promised individuals and pregnant
women
Split and subunit protein vaccines (natural or recombinant)
Downloaded by [University of Florida] at 02:10 04 December 2017

Non-infectious No or limited innate defence triggers Influenza (Fluarix, Fluarix Tetra,


Low reactogenicity Often have reduced immunogenicity Flulaval, Intanza, Vaxigrip, etc.),
compared to whole pathogen acellular pertussis, hepatitis B
vaccines (Engerix B, Recombivax HB),
For those vaccines with lower human papillomavirus (Cervarix,
immunogenicity, adjuvants are Gardasil, Gardasil 9), meningococ-
often needed cal B (Bexsero, Trumenba), malaria
(Mosquirix), herpes zoster
(Shingrix)
Toxoid vaccines
Vast experience as mature technology Vaccines only target the toxin and do Tetanus, diphtheria, acellular pertussis
not prevent infection by the (as part of DTaP combination vac-
pathogen cines: Boostrix, Infanrix, Adacel,
Non-infectious No herd protection etc.)
Used as carrier proteins due to good Priming and boosting necessary
immunogenicity
Polysaccharide vaccines
Easily identifiable target Weakly immunogenic, elicit only a Pneumococcal polysaccharide vaccine
transient antibody response giving (Pneumovax 23), meningococcal
a limited duration of protection polysaccharide vaccine (Mencevax)
Limited immunogenicity in infants
Hyporesponsiveness after repeated
doses
Limited or no impact on carriage
Polysaccharide conjugate vaccines
Improved memory responses leading Booster doses may be required to Meningococcal C (Neisvac), A
to increased protection in infants attain long-term protection (MenAfriVac), ACWY (Nimenrix,
Provide a longer duration of protec- Menveo, Menactra), pneumococcal
tion than polysaccharide vaccines conjugate vaccine (Prevnar,
due to B- and T-cell responses Prevnar 13, Synflorix), Haemophilus
Impact on carriage and transmission influenzae type b (ACT-HIB)
Reassortant live attenuated
Benefit from a live infection with a Contraindicated for immunocomprom- Rotavirus (RotaTeq)
non-pathogen strain ised individuals
Cannot cause the original disease, Risk of re-reassortants, especially
good safety profile when combination of several reas-
sortants [69]
Improved tolerability due to combin- Immunogenicity limited to selected
ation of low pathogenic virus with antigens
antigen from high pathogenic virus

OPV is a live attenuated vaccine that was obtained provides a high level of herd immunity through viral
through serial passages in non-human cells [19]. The shedding and can be quickly and easily imple-
attenuation of the three polio vaccine strains (Sabin 1, mented in outbreak settings. However, in very rare
2 and 3) greatly reduces their neurovirulence and cases (one case per million doses), OPV can mutate
transmissibility. OPV is easily administered through into a virulent form and induce very rare cases of
oral drops, inexpensive, and effective at inducing intes- vaccine-associated paralytic poliomyelitis [21]. As OPV
tinal mucosal immunity [20]. Furthermore, OPV has the rare capacity to become pathogenic again,
ANNALS OF MEDICINE 5

its use is incompatible with the final stages of polio require several doses or adjuvants to improve
eradication (i.e. eradication of both wild and vaccine- immunogenicity [27]. Therefore, these vaccines are
derived polio) and should be stopped after wild usually given repeatedly based on the prime-boost
poliovirus transmission has been controlled. For this principle to induce long-term immunity. This strat-
reason and to maintain population immunity, OPV egy, used since the advent of vaccines, aims at
has been replaced by an inactivated polio vaccine boosting antibody and cell-mediated immune
(IPV) in the United States, Europe, and an increasing responses through multiple primary doses and regu-
number of countries worldwide [22]. However, IPV is lar boosters. Impact on asymptomatic nasopharyn-
less effective than OPV at inducing intestinal muco- geal carriage of the pathogen and, consequently,
sal immunity among previously unvaccinated individ- herd protection by interrupting transmission can be
uals and is less effective at inducing herd immunity achieved with some non-live vaccines. Non-live vac-
via viral shedding [20]. Thus, as recently highlighted cines can contain inactivated whole pathogens or
by the World Health Organization, both vaccines only parts of them such as proteins or polysacchar-
(OPV first and then IPV) are crucial for the eradica- ides (subunit vaccines).
tion of polio [20].
Live attenuated vaccines targeting the same patho-
gen can also rely on different designs. This is the case 3.1. What are inactivated vaccines?
Downloaded by [University of Florida] at 02:10 04 December 2017

for vaccines against rotaviruses, a major cause of


Vaccines based on inactivated pathogens are pro-
acute gastroenteritis in children. Rotarix (GSK) is a sin-
duced by inactivating preparations of whole patho-
gle-strain human rotavirus live attenuated vaccine
gens by heat, radiation, or chemicals such as formalin
mimicking natural infection, whereas RotaTeq (MSD) is
or formaldehyde. Inactivation destroys the pathogen’s
a chimeric pentavalent human-bovine reassortant vac-
ability to replicate and cause the disease but maintains
cine, in which a nonpathogenic bovine rotavirus strain
its immunogenicity, so that the immune system can
provides the backbone to express the antigenic
still recognize the targeted pathogen.
proteins of the five most frequent human rotavirus
Inactivation approaches were first used to create
serotypes [23].
vaccines against pathogens such as typhoid fever,
Attenuation by cold-adaptation of the original
plague and cholera [1]. Current examples of inacti-
pathogen is used for live attenuated intranasal influ-
vated vaccines include the previously mentioned IPV,
enza vaccine (FluMist, Fluenz, MedImmune). It is pro-
whole-cell pertussis, rabies and hepatitis A vaccines
duced by inserting the genes of the antigenic
[1,28]. Whole-cell pertussis vaccines are produced
proteins from circulating influenza viruses in the
locally in many countries using different methods;
backbone of an attenuated cold-adapted influenza
thus, they are heterogeneous and may elicit different
virus. Intranasal administration of this vaccine mimics
immune responses [29]. In industrialized countries,
the natural route of influenza infection and induces
they have been frequently replaced by the less reacto-
an immune response directly in the respiratory
genic acellular vaccines based on a small number of
tract [24].
selected antigens of the pathogen.
The only live attenuated bacterial vaccine currently
in use is the bacillus Calmette-Gu erin (BCG) vaccine,
which was developed almost a century ago and is still
3.2. What are subunit vaccines?
the only licensed vaccine against tuberculosis [25].
This vaccine contains a bovine Mycobacterium bovis Subunit vaccines contain selected fragments of the
bacillus strain that had been attenuated by cell culture pathogen as antigens instead of the whole pathogen.
passage over several years [26]. These fragments can be proteins, polysaccharides, or
parts of a virus that may form virus-like particles
(VLPs). Subunit vaccines generally cause less adverse
3. Non-live vaccines
reactions than live or inactivated whole-organism vac-
Non-live vaccines do not contain any living or infec- cines, but they may be less immunogenic because
tious particles, so they cannot cause disease and they contain fewer antigens and the purification pro-
cannot reactivate. Therefore, they generally have a cess often eliminates components that trigger innate
good safety profile, even in immunocompromised immunity [30]. Examples of subunit vaccines include
individuals. However, a drawback of these vaccines tetanus toxoid, inactivated split and subunit seasonal
is that immunogenicity and duration of protection influenza, acellular pertussis and pneumococcal poly-
tend to be less than for live vaccines, and they may saccharide vaccines.
6 V. VETTER ET AL.

3.3. What are the different types of subunit The concept of combining recombinant proteins
vaccines? helped to develop the first malaria vaccine (Mosquirix,
GSK). In this vaccine, the gene of a surface protein of
3.3.1. Protein vaccines
the infectious form of Plasmodium falciparum is fused
Antigenic proteins can be purified from preparations to the HBsAg gene, and the resulting recombinant
of the whole pathogen, as for the acellular pertussis fusion protein is expressed in yeast with free recom-
vaccines [31], or can be produced by recombinant binant HBsAg [37]. These proteins spontaneously
genetic engineering [32]. In the latter case, a gene assemble into non-infectious VLPs (see Section 3.3.3)
encoding the antigenic protein is inserted into an and are combined with an Adjuvant System (AS) to
expression system able to produce large quantities of allow protection. The vaccine induces antibodies
the antigen in cell cultures. against parasites that have reached or are in transit to
Since the 1970s, most influenza vaccines have con- the liver (where they mature and multiply), thereby
sisted of split viruses deriving from preparation of limiting the ability of the parasites to cause clinical dis-
inactivated influenza vaccines. Inactivated influenza ease. Due to the high frequency of malaria in children
vaccine production is a long process relying on in some parts of the world, this vaccine has the poten-
embryonated chicken eggs, except for Flucelvax tial to prevent a large number of malaria cases [37,38].
(Seqirus), which is grown in mammalian cell culture The advantages of protein vaccines have been used
Downloaded by [University of Florida] at 02:10 04 December 2017

[33]. These split vaccines are then prepared by chem- to develop a new subunit vaccine against herpes zos-
ical disruption of the viral lipid envelope and, there- ter (Shingrix, GSK). This vaccine, containing a recom-
fore, the viral particle organization. These vaccines binant varicella-zoster virus protein and an AS, has the
retain immunogenicity but induce fewer adverse reac- objective of maintaining high efficacy also in older
tions than whole-virus inactivated influenza vaccines. adults who are at higher risk of herpes zoster [39,40].
Seasonal influenza vaccines contain in general three or It is also being studied for use in immunocomprom-
four viral strains to cover the influenza A and B strains ised individuals for whom live attenuated vaccines are
predicted to circulate during the forthcoming influenza often not recommended.
season. A good match between the vaccine strains Finally, reverse vaccinology is a new technology in
and the circulating strains is crucial to achieve high which genes encoding potential antigenic proteins are
protection rates [34]. Influenza vaccination is therefore identified from the entire genome of a given pathogen
recommended annually because the viruses in circula- [41]. The identified proteins are then tested in vitro and
tion frequently change and because protective immun- in vivo to determine whether they are immunogenic
ity is of short duration. and induce protective antibodies. Reverse vaccinology
Acellular pertussis vaccines are other examples of has been used to develop a vaccine against the chal-
purified antigenic proteins. These vaccines contain lenging Neisseria meningitidis serogroup B [42]. Unlike
between one and five highly purified pertussis anti- other N. meningitidis serogroups, serogroup B is cov-
gens, compared to more than 3000 antigens for ered by capsular polysaccharides that have similarities
whole-cell inactivated pertussis vaccines [31,35]. They to human polysaccharides. This property substantially
have been introduced in many industrialized countries reduces the immunogenicity of these polysaccharides
after concerns about whole-cell pertussis tolerance and could, at least theoretically, trigger antibodies
profile arose in the population, leading to lower vac-
against the human host and cause auto-immune dis-
cination coverage. Recently, a resurgence of pertussis
eases [42]. In addition, a vaccine relying on recombin-
has been observed in many regions of the world des-
ant proteins has been unsuccessful because of the high
pite high acellular pertussis vaccine coverage.
antigenic variation in circulating strains [42]. Reverse
Although the reasons for this resurgence are complex,
vaccinology helped identify four novel antigenic pro-
the shorter duration of protection and the probable
teins, which have been combined in a tetravalent men-
lower impact of these vaccines on infection and trans-
ingococcal B vaccine (Bexsero, GSK) [42]. By contrast, a
mission might play a role [31].
more traditional approach of protein screening was
An example of recombinant protein vaccine is pro-
used to develop a bivalent meningococcal B vaccine
vided by the widely used hepatitis B vaccine in which
(Trumenba, Pfizer) [43].
the gene of the hepatitis B surface antigen (HBsAg)
has been inserted into appropriate vectors for produc-
3.3.2. Toxoid vaccines
tion in yeast (Engerix-B, GSK; Recombivax-HB, MSD) or
mammalian cells (GenHevac-B, Sanofi Pasteur) [36]. Some bacteria such as Clostridium tetani, Clostridium
The resulting recombinant protein is then purified. difficile or Corynebacterium diphtheriae cause disease
ANNALS OF MEDICINE 7

by releasing pathogenic toxins. Vaccines against these during disease pathogenesis [47]. First-generation vac-
diseases are produced by detoxifying the toxin using cines against these pathogens were based on capsular
heat, chemicals (e.g. formaldehyde) or both. The inacti- polysaccharides purified from whole pathogens, such
vated toxins, called toxoids, are no longer pathogenic as the 23-valent pneumococcal polysaccharide vaccine
but retain their ability to induce toxin-neutralizing that was licensed in 1983 (Pneumovax 23, MSD;
antibodies. Classical examples of toxoid vaccines are Pneumo 23, Sanofi Pasteur).
those against diphtheria and tetanus, which have been However, polysaccharide vaccines are poorly
used since their discovery in the 1920s [44–46]. immunogenic, provide only short term protection, and
Pertussis toxoid is also included in all acellular pertus- can lead to a reduced immune response after repeated
sis vaccines [31]. Due to their good immunogenicity, vaccinations (i.e. hyporesponsiveness) [51,52]. Indeed,
toxoids are also used as carrier proteins for conjugate capsular polysaccharides are large molecules with
vaccines (see section 3.3.5) [47,48]. repeated antigens that can be recognized by B cells
Toxoids provide protection through the induction leading to their direct activation without the need of
of antibodies that must be present at disease onset to T-cell help. Furthermore, polysaccharide vaccines are
be effective. For this reason, toxoid vaccines require largely ineffective in children <2 years of age who are
multiple doses to maintain adequate life-long protec- at high risk for invasive disease by these pathogens
tion. However, toxoids protect only against disease due to the immaturity of their immune system [51].
Downloaded by [University of Florida] at 02:10 04 December 2017

pathogenesis in vaccinated individuals but do not pre-


vent infection or transmission [49]. Consequently, all 3.3.5. Polysaccharide conjugate vaccines
individuals need to be vaccinated regularly, and indir-
ect protection of unvaccinated people is generally not Studies performed in the 1920s–1930s showed that
possible. This is notably illustrated by the tetanus vac- the immunogenicity of purified polysaccharides could
cine. Tetanus is not transmitted from person-to-person be enhanced by coupling (i.e. conjugating) them to a
but occurs through contamination of wounds with protein [47]. This breakthrough allowed the develop-
C. tetani spores that are widespread in the environ- ment of second-generation vaccines against H. influen-
ment such as in the soil [45]. Therefore, high vaccin- zae type b, S. pneumoniae and N. meningitidis
ation coverage does not provide herd protection and serogroups A, C, W and Y that are effective in infants
unvaccinated or individuals not receiving regular [47]. Conjugation transforms the T-cell-independent
booster doses are potentially at risk. response induced by polysaccharides into a
T-cell-dependent response that induces high-affinity
3.3.3. VLPs antibodies and immune memory [47]. In addition, in
contrast to polysaccharide vaccines, conjugate
VLP vaccines are based on the observation that vaccines can induce herd protection by reducing
expression of certain viral proteins leads to the spon- asymptomatic nasopharyngeal carriage and, therefore,
taneous assembly of particles structurally similar to the transmission of these pathogens [47].
original viruses [50]. VLPs are not infectious because Conjugate vaccines are produced by chemically
they lack the viral genome. However, the native con-
linking polysaccharides to a carrier protein, which
formation of the antigenic proteins is well preserved,
makes the production of these vaccines more com-
which improves their immunogenicity compared to
plex. Different carrier proteins have been used, such as
free proteins.
tetanus and diphtheria toxoids, the nontoxic variant of
The best-known examples of VLPs are the human
diphtheria toxin CRM197 (isolated from C. diphtheriae
papillomavirus (HPV) vaccines that protect against cer-
C7 [b197] cultures), the outer membrane protein com-
vical cancer caused by these oncogenic viruses [50].
plex from N. meningitidis, or non-typeable H. influenzae
Currently, licensed HPV vaccines contain antigenic
protein D. The nature of the carrier protein and the
proteins from HPV types responsible for most cervical
chemical methods for conjugation may influence
cancers (e.g. Cervarix, GSK; Gardasil 9, MSD).
immunogenicity of the vaccines [47,48,53,54].
3.3.4. Polysaccharide vaccines
4. What are adjuvants?
Streptococcus pneumoniae, Haemophilus influenzae type
b and N. meningitidis are three encapsulated bacteria Adjuvants are substances that can enhance and modu-
that cause severe invasive disease. They possess poly- late the immunogenicity of the antigen [55]. Adjuvants
saccharide capsules that facilitate bacteria’s survival are usually not needed for live attenuated vaccines
when carried in the nasopharynx and in the blood because these vaccines actively replicate and
8 V. VETTER ET AL.

self-enhance the immune response. However, they are Vectored vaccines combine the advantages of live
frequently used for subunit vaccines because these vaccines and subunit vaccines and are made from
vaccines contain fewer antigens and lack some of the non-pathogenic infectious viruses expressing antigenic
intrinsic components present in whole pathogens that protein genes of a pathogen. Viral vectors derived
trigger the innate immune response, so that an effect- from retroviruses, herpes simplex viruses, adenoviruses
ive downstream adaptive response is less likely to be or poxviruses have been developed for vaccination
achieved [55]. against a wide array of pathogens, some of which
Due to their capacity to activate innate immune have been evaluated in clinical trials over the last few
responses, adjuvants can broaden or extend responses years [59]. However, their use can be limited by pre-
and improve memory responses, therefore allowing to exposure to the viruses used as vectors, leading to
reduce the number of doses needed or the amount of high prevalence of pre-existing neutralizing antibodies
antigen needed in each dose (dose sparing) [55]. against the vectors in humans, which can lead to early
These features can have important implications for vaccine clearance and reduced immunogenicity [59].
improving global vaccine supply, as illustrated during Another promising approach is the development of
the 2009–2010 influenza H1N1 pandemic. Adjuvants nucleic acid-based vaccines. These vaccines work by
can also improve immune responses in populations inserting DNA or RNA that encode antigenic proteins
into body cells (e.g. muscle or skin cells), which indu-
Downloaded by [University of Florida] at 02:10 04 December 2017

having typically low responses (e.g. infants, older


ces antigen presentation to the immune system trig-
adults or immunocompromised individuals) [55].
gering an immune response [60,61].
For almost a century, aluminium salts (also known
In addition to the conventional needle injections
as alum) were the only adjuvant approved worldwide
(intramuscular, subcutaneous and intradermal routes),
and they still remain the most widely used. Aluminium
alternative modes of vaccine delivery are also being
salts act primarily by directly activating innate immune
developed [62]. These include administration through
cells leading to antibody production [56]. Although
mucosal tissues (intranasal, oral or sublingual) or
many adjuvants have been developed, only a few
through the skin using microneedle or needle-free
have reached licensure stage due to the necessity of
devices. Vaccination through these alternatives routes
finding the right match between the antigen and the
is likely to be easier and more comfortable for vaccine
adjuvant: MPL (a detoxified form of bacterial lipopoly-
recipients and may improve vaccine acceptance and
saccharide), oil-in-water emulsions (MF-59), the AS
uptake, especially in those who fear needles [63,64].
combinations (AS01–04) and virosomes are currently
Furthermore, they can induce both systemic and
approved for humans. Virosomes consist of spherical
mucosal immune responses at the sites of pathogen
lipid layers assembled in vitro with viral proteins to
entry [62]. However, despite much effort, vaccination
resemble viral membranes [57]. They are currently
through these alternative routes remains limited to a
used in influenza and hepatitis A vaccines.
few vaccines (OPV, cholera vaccine, and rotavirus vac-
Adjuvants may be combined to obtain the desired
cines for the oral route and the live attenuated influ-
immune response, notably when cell-mediated
enza vaccine for the nasal route).
immune responses are needed. For instance, AS03 is
based on a-tocopherol (vitamin E) and squalene in an
oil-in-water emulsion, whereas AS04 is based on MPL 6. Are vaccines safe and always effective?
and aluminium salts [58]. AS03 is used in pandemic
Like all medicines, vaccines can have adverse events.
influenza vaccines (Pandemrix, Arepanrix, Adjupanrix,
However, because vaccines are given as preventive
GSK) and AS04 is used in a hepatitis B virus vaccine
measures mostly to healthy individuals, especially
(Fendrix, GSK) and a HPV vaccine (Cervarix, GSK).
infants and children, a highly positive benefit–risk pro-
file is essential. Vaccine safety is evaluated in the pre-
clinical and clinical phases of development but is also
5. Vaccines of the future
continuously monitored after licensure. Surveillance of
New vaccine designs and concepts are needed to vaccination programs and reporting by healthcare pro-
improve existing vaccines or address unmet needs viders are essential to detecting rare or serious adverse
notably for pathogens with multiple serotypes (e.g. events linked to vaccination, such as intussusception
dengue, S. pneumoniae), antigenic hypervariability (e.g. with the first rotavirus vaccine Rotashield (Wyeth) or
human immunodeficiency virus) or an intracellular narcolepsy with the adjuvanted H1N1 influenza vac-
phase that are predominantly controlled by T-cell cine Pandemrix (GSK) [23,65]. However, further
responses (e.g. tuberculosis, malaria) [11]. research is needed to confirm what role Pandemrix
ANNALS OF MEDICINE 9

may have played in the development of narcolepsy outside of this submitted work and is an advisory board
among those affected. Indeed, narcolepsy is a complex member for the GSK group of companies, Mithra, Pfizer and
disease whose causes are not yet fully understood but Amgen outside this submitted work.
Adjupanrix, Arepanrix, Bexsero, Boostrix, Cervarix, Engerix
it may be associated with genetic and environmental B, Fendrix, Flulaval, Havrix, Infanrix, Ixiaro, Mencevax, Menveo,
factors, including infections [66]. Mosquirix, Nimenrix, Pandemrix, Priorix, Priorix-Tetra, Rotarix,
Despite the recent successful developments in vac- Synflorix, Shingrix and Tritanrix are trademarks owned by or
cine design, no vaccine provides an absolute or life- licensed to the GSK group of companies. Gardasil, Gardasil 9,
long protection for all vaccinated individuals. In some M-M-RVAXPRO, Pneumovax 23, ProQuad, Recombivax HB,
cases, vaccines fail to induce a protective immune RotaTeq, Vaqta and Zostavax are trademarks of MSD (Merck
Sharp & Dohme) or Merck & Co, Inc. ACT-HIB, Adacel,
response. These vaccine failures are illustrated by
Dengvaxia, GenHevac-B, Imojev, Menactra, Pneumo 23,
breakthrough cases or are detected through sero- Stamaril and Vaxigrip are trademarks of Sanofi Pasteur.
logical testing of high-risk populations (e.g. hepatitis B Neisvac, Prevnar, Prevnar 13 and Trumenba are trademarks of
in healthcare providers, rubella in pregnant women). Pfizer. Fluenz and Flumist are trademarks of MedImmune, LLC.
For this reason, other available preventive measures Flucelvax is a trademark of Seqirus. Rotavac is a trademark of
should not be neglected, such as regular cervical Bharat Biotech. Dukoral is a trademark of Valneva Sweden AB.
screenings in women vaccinated against HPV [67].
Downloaded by [University of Florida] at 02:10 04 December 2017

Funding
7. Conclusions This work was supported by GlaxoSmithKline Biologicals S.A.,
Our improved understanding of the immune system which paid for all costs associated with the development
and host-pathogen interactions has allowed transition and the publishing of the present manuscript.
from an empirical to a more rational vaccine design, but
progress is still needed to address unmet needs and ORCID
improve protection induced by current vaccines.
Volker Vetter http://orcid.org/0000-0002-8414-1657
Vaccines with a similar indication can rely on very differ- Leonard R. Friedland http://orcid.org/0000-0001-9588-
ent concepts with their own benefits and limitations. 2306
Understanding the basic concepts of vaccines and their
recommendations for use is therefore crucial to under-
stand their benefits and risks. Notably, the number of
References
cases and deaths from infectious diseases prevented by
direct and indirect effects of vaccination worldwide and [1] Delany I, Rappuoli R, De Gregorio E. Vaccines for the
the economic benefit to societies by cost of prevention 21st century. EMBO Mol Med. 2014;6:708–720.
[2] Tognotti E. The eradication of smallpox, a success story
should be acknowledged. Improving public knowledge
for modern medicine and public health: what lessons
about vaccines should support the appropriate expecta- for the future? J Infect Dev Ctries. 2010;4:264–266.
tions of vaccines, increase confidence in the science of [3] Siegrist C-A. Vaccine immunology. In: Plotkin SA,
vaccination, and can help reduce vaccine hesitancy. Orenstein WA, Offit PA, editors. Vaccines. 6th ed.
Philadelphia, United States: Elsevier/Saunders; 2013.
p. 14–32.
Acknowledgements
[4] Clem AS. Fundamentals of vaccine immunology.
The authors thank Drs. Lode Schuerman and Alberta Di J Glob Infect Dis. 2011;3:73–78.
Pasquale (GSK, Belgium) for their critical review of the manu- [5] Kim TH, Johnstone J, Loeb M. Vaccine herd effect.
script and Sarah Brown (Fishawack, United Kingdom) for the Scand J Infect Dis. 2011;43:683–689.
figure. Writing assistance was provided by Dr. Julie Harriague [6] Fine PE. Herd immunity: history, theory, practice.
(4Clinics, France), on behalf of GSK. Authors would like to Epidemiol Rev. 1993;15:265–302.
thank Business & Decision Life Sciences platform for editorial [7] Orenstein W, Seib K. Mounting a good offense against
assistance and manuscript coordination, on behalf of GSK. measles. N Engl J Med. 2014;371:1661–1663.
Carole Desiron coordinated manuscript development and [8] Swamy GK, Heine RP. Vaccinations for pregnant
editorial support. women. Obstet Gynecol. 2015;125:212–226.
[9] Paterson P, Meurice F, Stanberry LR, et al. Vaccine
hesitancy and healthcare providers. Vaccine.
Disclosure statement
2016;34:6700–6706.
VV, LRF and JK are employees of the GSK group of compa- [10] Hajj Hussein I, Chams N, Chams S, et al. Vaccines
nies and hold shares in the GSK group of companies. GD is through centuries: major cornerstones of global
an employee of MSD and was an employee of the GSK health. Front Public Health. 2015;3:269.
group of companies. MS has received speaker honoraria [11] Pulendran B, Ahmed R. Immunological mechanisms of
from Merck, Amgen, Pfizer, NovoNordisk and Allergan vaccination. Nat Immunol. 2011;12:509–517.
10 V. VETTER ET AL.

[12] World Health Organization. Rubella vaccines: WHO [32] Clark TG, Cassidy-Hanley D. Recombinant subunit vac-
position paper. Wkly Epidemiol Rec. 2011;86:301–316. cines: potentials and constraints. Dev Biol (Basel).
[13] Monath TP, Gershman M, Staples JE, et al. Yellow 2005;121:153–163.
fever vaccine. In: Plotkin SA, Orenstein WA, Offit PA, [33] Soema PC, Kompier R, Amorij JP, et al. Current and
editors. Vaccines. 6th ed. Philadelphia, United States: next generation influenza vaccines: formulation and
Elsevier/Saunders; 2013. p. 870–968. production strategies. Eur J Pharm Biopharm. 2015;
[14] Yun SI, Lee YM. Japanese encephalitis: the virus and 94:251–263.
vaccines. Hum Vaccin Immunother. 2014;10:263–279. [34] World Health Organization. Vaccines against influenza
[15] Guy B, Briand O, Lang J, et al. Development of the WHO position paper – November 2012. Wkly
Sanofi Pasteur tetravalent dengue vaccine: one more Epidemiol Rec. 2012;87:461–476.
step forward. Vaccine. 2015;33:7100–7111. [35] Offit PA, Quarles J, Gerber MA, et al. Addressing
[16] Tillieux SL, Halsey WS, Sathe GM, et al. Comparative parents' concerns: do multiple vaccines overwhelm or
analysis of the complete nucleotide sequences of weaken the infant's immune system? Pediatrics.
measles, mumps, and rubella strain genomes con- 2002;109:124–129.
tained in Priorix-Tetra and ProQuad live attenuated [36] Michel ML, Tiollais P. Hepatitis B vaccines: protective
combined vaccines. Vaccine. 2009;27:2265–2273. efficacy and therapeutic potential. Pathol Biol (Paris).
[17] Tillieux SL, Halsey WS, Thomas ES, et al. Complete 2010;58:288–295.
DNA sequences of two oka strain varicella-zoster virus [37] Kaslow DC, Biernaux S. RTS,S: toward a first landmark
genomes. J Virol. 2008;82:11023–11044. on the Malaria Vaccine Technology Roadmap. Vaccine.
Downloaded by [University of Florida] at 02:10 04 December 2017

[18] Harpaz R, Ortega-Sanchez IR, Seward JF. Prevention of 2015;33:7425–7432.


herpes zoster: recommendations of the Advisory [38] Vandoolaeghe P, Schuerman L. The RTS,S/AS01 mal-
Committee on Immunization Practices (ACIP). MMWR aria vaccine in children 5 to 17 months of age at first
Recomm Rep. 2008;57:1–30. quiz CE2–4. vaccination. Expert Rev Vaccines. 2016;15:1481–1493.
[19] Sabin AB, Boulger LR. History of Sabin attenuated [39] Lal H, Cunningham AL, Godeaux O, et al. Efficacy of
poliovirus oral live vaccine strains. J Biol Stand. an adjuvanted herpes zoster subunit vaccine in older
1973;1:115–118. adults. N Engl J Med. 2015;372:2087–2096.
[20] World Health Organization. Polio vaccines: WHO pos- [40] Cunningham AL, Lal H, Kovac M, et al. Efficacy of the
ition paper – March, 2016. Wkly Epidemiol Rec. herpes zoster subunit vaccine in adults 70 years of
2016;91:145–168. age or older. N Engl J Med. 2016;375:1019–1032.
[21] Esteves K. Safety of oral poliomyelitis vaccine: results [41] Seib KL, Zhao X, Rappuoli R. Developing vaccines in
of a WHO enquiry. Bull World Health Org. the era of genomics: a decade of reverse vaccinology.
1988;66:739–746. Clin Microbiol Infect. 2012;18 Suppl 5:109–116.
Vernikos G, Medini D. BexseroV chronicle. Pathog
R
[22] Doherty M, Buchy P, Standaert B, et al. Vaccine [42]
impact: benefits for human health. Vaccine. 2016;34: Glob Health. 2014;108:305–316.
6707–6714. [43] Shirley M, Dhillon S. Bivalent rLP2086 Vaccine
(TrumenbaV): a review in active immunization against
R
[23] Vesikari T. Rotavirus vaccination: a concise review. Clin
Microbiol Infect. 2012;18(Suppl 5):57–63. invasive Meningococcal Group B disease in individuals
[24] Sridhar S, Brokstad KA, Cox RJ. Influenza vaccination aged 10–25 years. BioDrugs. 2015;29:353–361.
strategies: comparing inactivated and live attenuated [44] Moloney PJ. The preparation and testing of diphtheria
influenza vaccines. Vaccines (Basel). 2015;3:373–389. toxoid (Anatoxine-Ramon). Am J Public Health (NY).
[25] Principi N, Esposito S. The present and future of 1926;16:1208–1210.
tuberculosis vaccinations. Tuberculosis (Edinb). [45] World Health Organization. Tetanus vaccine. WHO
2015;95:6–13. position paper. Wkly Epidemiol Rec. 2006;81:198–208.
[26] World Health Organization. BCG vaccine. WHO pos- [46] World Health Organization. Diphtheria vaccines. WHO
ition paper. Wkly Epidemiol Rec. 2004;79:27–38. position paper. Wkly Epidemiol Rec. 2006;81:21–32.
[27] Strugnell R, Zepp F, Cunningham A, et al. Vaccine anti- [47] Pichichero ME. Protein carriers of conjugate vaccines:
gens. In: Garçon N, Stern PL, Cunningham AL, editors. characteristics, development, and clinical trials. Hum
Understanding modern vaccines: perspectives in vacci- Vaccin Immunother. 2013;9:2505–2523.
nology. Vol. 1. Amsterdam: Elsevier; 2011. p. 61–88. [48] €ker M, Berti F, Schneider J, et al. Polysaccharide
Bro
[28] Cunningham AL, Garcon N, Leo O, et al. Vaccine conjugate vaccine protein carriers as a “neglected
development: from concept to early clinical testing. valency” – potential and limitations. Vaccine. 2017;35:
Vaccine. 2016;34:6655–6664. 3286–3294.
[29] Edwards KM, Decker MD. Pertussis vaccines. In: Plotkin [49] Borba RC, Vidal VM, Moreira LO. The re-emergency
SA, Orenstein WA, Offit PA, editors. Vaccines. 6th ed. and persistence of vaccine preventable diseases. An
Philadelphia, United States: Elsevier/Saunders; 2013. Acad Bras Cienc. 2015;87:1311–1322.
p. 447–492. [50] Roldao A, Mellado MC, Castilho LR, et al. Virus-like
[30] Rappuoli R, Pizza M, Del Giudice G, et al. Vaccines, particles in vaccine development. Expert Rev Vaccines.
new opportunities for a new society. Proc Natl Acad 2010;9:1149–1176.
Sci USA. 2014;111:12288–12293. [51] Hutter J, Lepenies B. Carbohydrate-based vaccines: an
[31] World Health Organization. Pertussis vaccines: WHO overview. Methods Mol Biol. 2015;1331:1–10.
position paper – September 2015. Wkly Epidemiol [52] Poolman J, Borrow R. Hyporesponsiveness and its clin-
Rec. 2015;90:433–458. ical implications after vaccination with polysaccharide
ANNALS OF MEDICINE 11

or glycoconjugate vaccines. Expert Rev Vaccines. [62] Stanberry LR, Strugnell R. Vaccines of the future. In:
2011;10:307–322. Garçon N, Stern PL, Cunningham AL, editors.
[53] Kim JS, Laskowich ER, Arumugham RG, et al. Understanding modern vaccines: perspectives in vacci-
Determination of saccharide content in pneumococcal nology. Vol. 1. Amsterdam: Elsevier; 2011. p. 151–199.
polysaccharides and conjugate vaccines by GC-MSD. [63] Shakya AK, Chowdhury MYE, Tao W, et al.
Anal Biochem. 2005;347:262–274. Mucosal vaccine delivery: current state and a
[54] Poolman J, Frasch C, Nurkka A, et al. Impact of the pediatric perspective. J Control Release. 2016;240:
conjugation method on the immunogenicity of 394–413.
Streptococcus pneumoniae serotype 19F polysacchar- [64] Arnou R, Frank M, Hagel T, et al. Willingness to vac-
ide in conjugate vaccines. Clin Vaccine Immunol. cinate or get vaccinated with an intradermal seasonal
2011;18:327–336. influenza vaccine: a survey of general practitioners
[55] Pasquale AD, Preiss S, Silva FT, et al. Vaccine adju- and the general public in France and Germany. Adv
vants: from 1920 to 2015 and beyond. Vaccines Ther. 2011;28:555–565.
(Basel). 2015;3:320–343. [65] Sarkanen TO, Alakuijala APE, Dauvilliers YA, et al.
[56] Wen Y, Shi Y. Alum: an old dog with new tricks. Incidence of narcolepsy after H1N1 influenza and vac-
Emerg Microbes Infect. 2016;5:e25. cinations: systematic review and meta-analysis. Sleep
[57] Moser C, Muller M, Kaeser MD, et al. Influenza viro- Med Rev. 2017 [Jun 20]. DOI:10.1016/
somes as vaccine adjuvant and carrier system. Expert j.smrv.2017.06.006
Rev Vaccines. 2013;12:779–791. [66] Kornum BR, Knudsen S, Ollila HM, et al. Narcolepsy.
[58] Garcon N, Di Pasquale A. From discovery to licensure, Nat Rev Dis Primers. 2017;3:16100.
Downloaded by [University of Florida] at 02:10 04 December 2017

the adjuvant system story. Hum Vaccin Immunother. [67] Lee LY, Garland SM. Human papillomavirus
2017;13:19–33. vaccination: the population impact. F1000Res. 2017;
[59] Ramezanpour B, Haan I, Osterhaus A, et al. Vector- 6:866.
based genetically modified vaccines: exploiting [68] Ellis RW, Rappuoli R, Ahmed S. Technologies for mak-
Jenner's legacy. Vaccine. 2016;34:6436–6448. ing new vaccines. In: Plotkin SA, Orenstein WA, Offit
[60] Khan KH. DNA vaccines: roles against diseases. Germs. PA, editors. Vaccines. 6th ed. Philadelphia, United
2013;3:26–35. States: Elsevier/Saunders; 2013. p. 1182–1199.
[61] Deering RP, Kommareddy S, Ulmer JB, et al. Nucleic [69] Hemming M, Vesikari T. Detection of rotateq vaccine-
acid vaccines: prospects for non-viral delivery of derived, double-reassortant rotavirus in a 7-year-old
mRNA vaccines. Expert Opin Drug Deliv. 2014;11: child with acute gastroenteritis. Pediatr Infect Dis J.
885–899. 2014;33:655–656.

You might also like