Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jan 26.
Published in final edited form as: Curr Top Microbiol Immunol. 2020;428:1–30. doi: 10.1007/82_2018_102

Successful Vaccines

Ian J Amanna 1, Mark K Slifka 2
PMCID: PMC6777997  NIHMSID: NIHMS1052258  PMID: 30046984

Abstract

Vaccines are considered one of the most important advances in modern medicine and have greatly improved our quality of life by reducing or eliminating many serious infectious diseases. Successful vaccines have been developed against many of the most common human pathogens and this success has not been dependent upon any one specific class of vaccine since subunit vaccines, non-replicating whole-virus or whole-bacteria vaccines, and attenuated live vaccines have all been effective for particular vaccine targets. After completing the initial vaccination series, one common aspect of successful vaccines is that they induce long-term protective immunity. In contrast, several partially successful vaccines appear to induce protection that is relatively short-lived and it is likely that long-term protective immunity will be critical for making effective vaccines against our most challenging diseases such as AIDS and malaria.

1. Introduction

The effect of vaccines on public health is truly remarkable. One study examining the impact of childhood vaccination on the 2001 US birth cohort found that vaccines prevented 33,000 deaths and 14 million cases of disease (Zhou et al. 2005). Among 73 nations supported by the GAVI alliance, mathematical models project that vaccines will prevent 23.3 million deaths from 2011–2020 compared to what would have occurred if there were no vaccines available (Lee et al. 2013). Vaccines have been developed against a wide assortment of human pathogens (Table 1). There are vaccines against bacterial toxins (e.g., tetanus and diphtheria toxins), acute viral pathogens (e.g., measles, mumps, rubella), latent or chronic viral pathogens (e.g., varicella zoster virus [VZV] and human papilloma virus [HPV], respectively), respiratory pathogens (e.g., influenza, Bordetella pertussis), and enteric pathogens (e.g., poliovirus, Salmonella typhi). Most licensed vaccines can be categorized as (a) live, attenuated vaccines, (b) non-replicating whole-particle vaccines (including virus-like particles, or VLPs), and (c) subunit vaccines. There are similar numbers of licensed vaccines in each of these categories (Table 1) and this illustrates the point that no single vaccine approach is superior to another, but instead the approach varies according to each individual pathogen and the feasibility, safety, and efficacy required to develop a successful vaccine to that particular pathogen. Interestingly, influenza represents the only example in which there is at least one licensed vaccine listed in each of these three categories.

Table 1.

Licensed vaccines

Live, attenuated vaccines
Adenovirus, Types 4 and 7
Dengue virusa
Influenza virus
Measles virus
Mumps virus
Mycobacterium tuberculosis
Poliovirusb
Rotavirus
Rubella virus
Salmonella typhi (typhoid fever)
Varicella zoster virus (chickenpox and shingles)
Variola virus (smallpox)
Yellow fever virus
Non-Replicating microbial particle vaccines
Bordetella pertussisc (whooping cough)
Hepatitis A virus
Hepatitis B virus
Human papillomavirus
Influenza virusd
Japanese encephalitis virus
Neisseria meningitidis serogroup Be (bacterial meningitis)
Poliovirus
Rabies virus
Tick-borne encephalitis virusf
Yersinia pestisg (bubonic plague)
Subunit vaccines
Bacillus anthracis (anthrax)
Bordetella pertussis (whooping cough)
Diphtheria toxin
Haemophilus influenzae type b
Influenza virus
Neisseria meningitidis serogroups A, C, Y, W-135 and Be (bacterial meningitis)
Salmonella typhi (typhoid fever)
Streptococcus pneumoniae (pneumococcal disease)
Tetanus toxin
a

As of April 4, 2016 the Dengvaxia® live attenuated chimeric vaccine was licensed for use in Mexico, El Salvador, the Philippines and Brazil (Sanofi Pasteur 2016).

b

In the US, the oral poliovirus vaccine (OPV) was fully replaced by the inactivated poliovirus vaccine (IPV) by 2000 (Advisory Committee on Immunization Practices 2000).

c

The whole-cell pertussis (wP) vaccine was recommended to be fully replaced by the acellular pertussis (aP) vaccine by 1997 in the US, but continues to be used routinely in other countries (Advisory Committee on Immunization Practices 1997).

d

A whole-virion inactivated H5N1 pandemic influenza vaccine (Vepacel®) is licensed for use in the EU (Plosker 2012).

e

The Bexsero® meningococcal group B vaccine uses a combination of outer membrane vesicle particles as well as individual recombinant proteins (Novartis Vaccines and Diagnostics 2015).

f

A tick-borne encephalitis virus vaccine is not currently licensed for the US, but is used in multiple EU countries (Heinz et al. 2013).

g

Manufacture of the plague vaccine in the US was discontinued in 1999 (Williamson and Oyston 2013).

2. Successes and failures of live attenuated vaccines

There are currently 13 diseases for which live, attenuated vaccines have been developed and licensed for commercial use (Table 1). Development of successful vaccines is typically easier when there is (a) a single serotype, (b) the pathogen is antigenically stable, (c) diagnostic tests/clinical criteria for measuring disease burden are available, and (d) the vaccine is both safe and effective. Smallpox vaccination with vaccinia virus is the most famous example of a highly effective vaccine and at the time when people were faced with smallpox outbreaks, this vaccine was associated with each of these characteristics that led to the implementation of a successful vaccine. The smallpox vaccine is not only the first vaccine ever developed (Jenner 1798, 1799, 1800), but since Variola virus (the causative agent of smallpox) no longer exists in nature, this also represents the first example in which a vaccine was purposefully used to drive a species to extinction. Some of the best examples of successful live, attenuated vaccines include those developed against measles, mumps, and rubella (MMR). In contrast, other live, attenuated vaccines such as the oral polio vaccine (OPV) and the first licensed vaccine against rotavirus (RotaShield®) serve as important reminders that vaccines must be safe if they are going to be used successfully for routine vaccination.

2.1. Measles, Mumps, Rubella

One of the most common live attenuated vaccines in use today is the MMR vaccine. Although originally designed as three separate vaccines, attenuated strains of measles, mumps, and rubella were successfully formulated into a single combination vaccine in the US in 1971 (Strebel et al. 2013) and it continues to be used to provide protective immunity against pathogens responsible for what were once considered common childhood diseases. Similar to smallpox vaccination, they were developed against viruses that are antigenically stable and have only a single serotype. Even when combined with live, attenuated VZV to provide a quadrivalent vaccine, seroconversion rates remain high (measles; 97.1%, mumps; 96.0%, rubella; 98.8% and varicella; 93.5%, respectively) (Lieberman et al. 2006). Other vaccines against antigenically variable viruses (e.g., influenza) or pathogens with multiple serotypes (e.g., HPV) indicate that as vaccine technologies have improved and evolved, there are opportunities to combat more complicated and challenging pathogens through effective immunization programs. Although nearly all vaccines require at least one booster dose in order to maintain long-term protective immunity (Slifka and Amanna 2014), many of our most successful vaccines have achieved the appropriate balance of both safety and efficacy.

2.2. Rotavirus and Poliovirus

There are many aspects to a vaccine that are required for them to be considered successful and one of the most important parameters for routine vaccines is patient safety. When vaccines demonstrate safety and efficacy during Phase II/III trials, then they have the opportunity to be licensed for commercial production while continuing to be monitored post-licensure (i.e., Phase IV). The RotaShield vaccine developed against rotavirus represents a vaccine that failed post-licensure due to an unacceptably high rate of reactogenicity in which serious adverse events (e.g., intussusception) reached 90–214 cases per million administered doses (Kramarz et al. 2001; Murphy et al. 2001). Following licensure in 1998, it was subsequently pulled from the market by the end of 1999. Live, attenuated rotavirus vaccines with improved safety profiles (Rotarix® and RotaTeq®) have now entered the market to address this important clinical need. Similarly, live OPV provided good immunogenicity and protective efficacy, but also caused vaccine-associated paralytic poliomyelitis (VAPP) at a rate of 0.4–0.5 cases per million administered doses (Prevots et al. 1994; Hao et al. 2008). Because of its widespread use in the US, this resulted in an average of 9 children each year from 1961–1989 who suffered paralysis due to OPV (Alexander et al. 2004). Since polio was no longer endemic in the US, the OPV vaccine was causing more cases of paralysis than the wild-type polioviruses that it was designed to prevent. In 2000, the OPV vaccine regimen was abandoned in favor of the safer inactivated polio vaccine (IPV) series and VAPP has been eliminated (Alexander et al. 2004).

3. Successes and failures of non-replicating/inactivated vaccines

Successful non-replicating/inactivated vaccines have been licensed for 11 different pathogens and include both viruses and bacteria (Table 1). As described below, the hepatitis A virus (HAV) vaccine represents a striking success for whole-virion inactivated vaccines, with exceptional efficacy and long-term antibody persistence demonstrated for up to 20 years post-vaccination (Theeten et al. 2015). The HPV vaccine utilizes advanced VLP technology and is able to protect against chronic viral infection, prevent HPV-associated cancers, and potentially induce life-long immunity with as few as two doses or even a single immunization (Safaeian et al. 2013; Kreimer et al. 2015). In contrast to these success stories, unsuccessful attempts to develop formaldehyde-inactivated measles and respiratory syncytial virus (RSV) vaccines in the 1960s indicate that an inactivated whole-virus vaccine approach is not foolproof and several factors may have contributed to those failures (Polack 2007).

3.1. Hepatitis A

Infection with HAV was once a leading cause of viral hepatitis in the US, with 20,000–60,000 cases reported annually and fatality rates as high as 1.8% in adults >50 years of age (Centers for Disease Control and Prevention 2015c). Vaccine development against HAV benefited from several key factors including early studies demonstrating the efficacy of passive immunotherapy (Gellis et al. 1945; Havens and Paul 1945; Stokes and Neefe 1945; Hsia et al. 1954), the discovery of cell culture conditions for the propagation of HAV (Provost and Hilleman 1979), and passive and active immunization studies in appropriate animal models including chimpanzees and marmosets (Purcell et al. 1992; Provost et al. 1986). These critical elements supported the eventual development and licensure of two inactivated whole-virion, alum-adjuvanted vaccines (Havrix® and Vaqta®) in the US and since their implementation in the 1990s, the rates of disease have declined by >90% (Centers for Disease Control and Prevention 2015c). Efficacy for both vaccines is high, with estimates ranging from 94–100% (Werzberger et al. 1992; Innis et al. 1994). HAV-specific antibodies have been identified as the key correlate of protective immunity following vaccination, and although an absolute lower limit has not been defined, titers >10–20 mIU/mL are generally considered protective (Centers for Disease Control and Prevention 1996).

One of the most impressive aspects of the inactivated HAV vaccines has been the durability of vaccine-induced antibody responses. Preliminary studies examining HAV-specific antibody titers demonstrated a rapid drop shortly after vaccination, but also showed a second, slower rate of decay, with protective immunity estimated to be maintained for 20–30 years following vaccination (Van Damme et al. 1994). Longer-term studies have confirmed and extended these initial results (Wiedermann et al. 1997; Rendi-Wagner et al. 2007; Theeten et al. 2015). In one notable study, antibody titers in subjects who received 2 doses of inactivated HAV vaccine were followed annually for up to 20 years post-vaccination (Theeten et al. 2015). Based on these results, it was estimated that at 40 years post-vaccination, ≥90% of vaccinees would maintain antibodies above the putative protective threshold. This may actually be an underestimate since, as noted by the authors, antibody decline appeared to level off over time with minimal changes to average group antibody titers from 11–20 years post-vaccination (Theeten et al. 2015). Remarkably, this data indicates that two doses of an alum-adjuvanted inactivated vaccine may be sufficient to induce life-long immunity for most vaccine recipients.

3.2. Human Papilloma Virus

Exposure to HPV, typically transmitted at mucosal surfaces through sexual contact, can lead to persistent infection and a range of cancers (Centers for Disease Control and Prevention 2015d). Cervical cancer, caused almost exclusively by HPV, still bears a substantial burden on the US, with 4,074 deaths reported in 2012 alone (the most recent year with available data (Centers for Disease Control and Prevention 2015a)). Although more than 120 HPV types have been identified, a smaller subset of approximately 13–14 types are considered high-risk for developing cancer (Centers for Disease Control and Prevention 2015d). Successful development of HPV vaccines was precipitated by the discovery that the L1 capsid protein of the virus could self-assemble into VLPs and induce high-titer neutralizing antibodies (Kirnbauer et al. 1992). Papillomaviruses are species-restricted and this constrains the ability to assess HPV vaccine candidates directly in animals. However, several species-specific models (including mucosal challenge routes) were tested using L1 VLP candidates (Schiller and Lowy 1996). These animal studies demonstrated several key findings including (a) VLPs could protect prophylactically against disease but had no impact on established infections, (b) denatured VLPs were unable to elicit protective responses, suggesting that the three-dimensional conformation of the VLP was important to eliciting appropriate immunity, and (c) passively transferred antibodies from immunized animals were sufficient to protect against disease (Schiller and Lowy 1996).

These early animal studies of the L1 VLP approach were encouraging enough to prompt the NIH and several pharmaceutical companies to begin testing vaccine candidates in clinical studies (Schiller and Hidesheim 2000) that eventually led to key Phase III clinical trials with efficacy estimates ranging from 90–98% (Paavonen et al. 2007; The FUTURE II Study Group 2007). US licensure was granted by 2006 (quadrivalent Gardasil®) and 2009 (bivalent Cervarix®), with a 9-valent vaccine formulation (Gardasil 9®) licensed in 2014 (Centers for Disease Control and Prevention 2015d). Besides the number of HPV serotypes included in each formulation, the primary difference between the two vaccines is the inclusion of the monophosphoryl lipid A (MPL) adjuvant to the Cervarix vaccine in an effort to enhance antibody responses. In addition to providing impressive levels of protective efficacy, antibody responses to both vaccines appear to be long-lived. In one study, subjects vaccinated with 3-doses of either Cervarix or Gardasil were followed for up to 4 years (Einstein et al. 2014). While Cervarix induced higher serum and cervicovaginal neutralizing antibody titers, the rate of antibody decline was similar for both vaccines, with titers typically plateauing at 2–3 years and appearing stable thereafter (Einstein et al. 2014). Similar antibody kinetics have been observed by others (Giannini et al. 2006), with studies demonstrating both long-term antibody persistence (Safaeian et al. 2013) and efficacy following a single VLP vaccination (Kreimer et al. 2015). Although the precise mechanism of protection is unclear (Centers for Disease Control and Prevention 2015d), it is believed that serum neutralizing antibodies elicited by HPV vaccines can reach the genital tract through antibody exudation or transudation and thereby protect against infection at the mucosal surface (Schwarz and Leo 2008). Further evidence supporting this position comes from a recent study using a mouse cervicovaginal model which demonstrated that passive systemic transfer of immune serum alone could block HPV infection at the vaginal epithelium (Day et al. 2010). Unfortunately, despite proven efficacy, safety and durability of immunity, only 57% of adolescent girls and 35% of adolescent boys in the US had received a single HPV vaccine dose in 2013 (Stokley et al. 2014). This represents a significant missed opportunity, leaving a substantial number of young people at continued risk for infection and HPV-associated cancers.

3.3. Inactivated Measles and Respiratory Syncytial Virus Vaccines

The measles virus is one of the oldest recorded human pathogens, and despite the availability of an effective live attenuated vaccine, it remains a significant threat for many parts of the world (World Health Organization 2014). The first live vaccine was licensed in the US in 1963 and since that time the incidence of disease has dropped dramatically (Centers for Disease Control and Prevention 2015e). In addition to the live vaccine, a formaldehyde inactivated measles vaccine (FIMV) was also introduced in 1963 (Centers for Disease Control and Prevention 2015e). One trial involving over 5,000 children indicated the FIMV could induce seroconversion, though immunity appeared to wane rapidly with titers declining to undetectable levels within 6 months following vaccination (Carter et al. 1962). Efficacy studies reinforced this result, with a reasonably high efficacy of 81% observed during the first three months post-vaccination, but dropping to only 63% by 11–13 months (Guinee et al. 1966). More concerning however, were multiple reports detailing an unusual and severe form of measles following wild-type exposure of children, all having previously received the inactivated vaccine (Fulginiti et al. 1967; Rauh and Schmidt 1965). This condition, termed atypical measles, presented a range of symptoms including high rates of muscle and abdominal pain as well as peripheral edema and pneumonia (Fulginiti et al. 1967; Rauh and Schmidt 1965). Based on this and other reports, the vaccine was removed from the market by 1967, just 4 years after its initial licensure (Centers for Disease Control and Prevention 2015e).

RSV was first described in the 1950s, and was soon recognized as having a substantial impact on childhood respiratory health (Chanock et al. 1961; Beem et al. 1960). RSV remains a significant cause of acute lower respiratory infections among children <5 years of age, with a recent meta-analysis estimating an annual global burden of 33.8 million RSV-associated respiratory episodes and 66,000–199,000 deaths (Nair et al. 2010). Even in the US, an estimated 2.1 million children require medical attention each year due to RSV, with ~57,000 hospitalizations (Hall et al. 2009). Soon after the discovery of RSV, a formaldehyde-inactivated RSV (FIRSV) vaccine candidate was developed. However, multiple field efficacy trials demonstrated no protective immunity, and instead provided evidence for increased risk for enhanced RSV disease in vaccine recipients (Kapikian et al. 1969; Kim et al. 1969; Fulginiti et al. 1969; Chin et al. 1969). For example, children vaccinated with either the FIRSV candidate or an inactivated parainfluenza vaccine as a control, demonstrated RSV attack rates that were similar between these groups (65% and 53%, respectively). However, the hospitalization rates among infected children in the FISRV cohort reached 80% (compared to a rate of only 5% in the control group) and the vaccine was associated with 2 deaths (Kim et al. 1969).

The similarities between the failed measles virus and RSV inactivated vaccines are evident (Polack 2007). Both pathogens are respiratory infections belonging to the Paramyxoviridae family and while the underlying mechanism of immune enhancement of disease remains unclear, it is believed that inactivated vaccines against either virus fail to induce protective high-avidity antibody, with skewing towards a Th2 response and subsequent enhanced inflammation following wild-type infection (Polack 2007; Acosta et al. 2016). Importantly, it is also clear that neutralizing antibodies are key to controlling either disease. Passive immunotherapy for the prevention of measles was described as early as 1924 (Zingher 1924), with recent meta-analysis confirming its value (Young et al. 2014). Similarly, prophylactic administration of a RSV neutralizing monoclonal antibody (palivizumab) has been shown to improve clinical outcomes and reduce hospitalizations in high-risk infants across multiple trials (Feltes et al. 2003; Pedraz et al. 2003; The IMpact RSV Study Group 1998). These results suggest that protective immunity through vaccination is achievable for both pathogens, and this has been accomplished for measles through the use of a live attenuated vaccine for >50 years. In contrast, while several new RSV vaccine candidates are under development, none have reached licensure (Jaberolansar et al. 2016). Nevertheless, recent positive results with a recombinant RSV F protein vaccine candidate in women (Glenn et al. 2016) provides some encouragement that the field is moving forward.

4. Successes and failures of subunit vaccines

Subunit vaccines have been licensed for 9 pathogens and encompass a range of approaches including purified toxoids, recombinant proteins, and either free or protein-conjugated polysaccharides (Table 1). In this section, we highlight the historic successes achieved with tetanus and diphtheria toxoids, both of which were developed in the 1920s (Roper et al. 2013; Tiwari and Wharton 2013). As a more recent example, we explore the development and implementation of successful vaccines against Haemophilus influenza type B (Hib) (Briere et al. 2014). These three examples have all benefited from having a defined antigen target with mechanisms of immunity established through passive immunization studies in animal models as well as human clinical practice. By comparison, the development of acellular pertussis (aP) vaccines face ongoing challenges due to the waning immunity associated with this subunit vaccine approach (Plotkin 2014).

4.1. Tetanus and Diphtheria

Tetanus and diphtheria are both toxin-mediated diseases, against which effective vaccines have been widely available in the US for >70 years (Roper et al. 2013; Tiwari and Wharton 2013). Tetanus is mediated by the neurotoxin tetanospasmin (i.e., tetanus toxin) expressed by the bacterium, Clostridium tetani (Roper et al. 2013), and diphtheria results from the diphtheria toxin produced by Corynebacterium diphtheriae (Tiwari and Wharton 2013). Both vaccines consist of formaldehyde-inactivated toxoids with the diphtheria toxoid developed in 1920 (Tiwari and Wharton 2013) and the tetanus toxoid first described in 1924 (Roper et al. 2013). Efficacy of both toxoid vaccines has been demonstrated in large part by following epidemiology trends over time. Prior to broad introduction of the tetanus vaccine in the 1940s, the US recorded ~500–600 cases of tetanus annually (Centers for Disease Control and Prevention 2015f). In the post-vaccination era, this rate has dropped to ~30 cases annually and from 2001 – 2008 the CDC reported that in instances where vaccination status was reported, 41% of patients had never received a tetanus toxoid-containing vaccine (Centers for Disease Control and Prevention 2015f). Control of diphtheria (a communicable disease, in contrast to tetanus) has shown an even more dramatic impact. In the 1920s, there were an estimated 100,000–200,000 annual cases of diphtheria in the US with up to 15,000 deaths (Centers for Disease Control and Prevention 2015b). These rates dropped rapidly after introduction of the diphtheria vaccine, with no cases reported over an 11-year period from 2003–2013 (Adams et al. 2015). Immunity for both diseases is achieved through antibody-mediated toxin neutralization. For tetanus, immunity is considered 100% effective in preventing death if pre-existing antibody levels are above a putative protective threshold of 0.01 IU/mL (Amanna et al. 2008). This protective threshold is bolstered both by direct human challenge studies performed in the 1940s (Wolters and Dehmel 1942) as well as analysis of patients admitted with clinical signs of tetanus, wherein only those patients with titers <0.01 IU/mL experience the most severe symptoms or die from the disease (Goulon et al. 1972). For diphtheria, a minimal protective serum titer has also been defined as 0.01 IU/mL (Tiwari and Wharton 2013; Bjorkholm et al. 1986),

The success of the tetanus and diphtheria subunit vaccines may be viewed as a combination of several factors. Research into both toxins benefited from the development of relevant animal models and the demonstration that passive immunotherapy, directed against either toxin, was sufficient to achieve protective immunity (von Behring and Kitasato 1890). These results presented clear targets for vaccination with known mechanisms of protection (i.e., neutralizing antibodies). Furthermore, protection against both diseases can be achieved with relatively low levels of toxin-specific antibodies compared to the high levels of antibodies elicited through current vaccination schedules. For instance, following the completion of the primary tetanus and diphtheria vaccination series currently recommended in the US (consisting of 5 doses through 4–6 years of age) average anti-tetanus and anti-diphtheria antibody titers reach >7 IU/mL (Black et al. 2006), several orders of magnitude above their protective thresholds of 0.01 IU/mL. In terms of longevity, we and others have shown that while antibody titers decline rapidly during the first few years following vaccination (typically 1–3 years following immunization (Amanna and Slifka 2010; Embree et al. 2015)), anti-toxin antibodies then display longer-lived single-order kinetics, with tetanus-specific antibody half-lives ranging from 11 to 14 years (Amanna et al. 2007; Hammarlund et al. 2016; Bonsignori et al. 2009), and diphtheria-specific titers declining with an estimated half-life of 19 to 27 years (Amanna et al. 2007; Hammarlund et al. 2016). While these rates of decline are somewhat faster compared to other vaccine and viral antigens (Amanna et al. 2007), based on current antibody levels measured among US adults, at least 95% of the population will remain protected against both tetanus and diphtheria for 30 years or more without requiring further booster vaccination (Hammarlund et al. 2016). In light of these results, the current US recommendation for adult tetanus/diphtheria booster doses to be administered every 10 years (Kim et al. 2016) should be reconsidered.

4.2. Haemophilus Influenza Type B

Hib is a gram-negative bacterium that was once the leading cause of bacterial meningitis in US children (Briere et al. 2014). In the pre-vaccine era, annual US rates of invasive Hib in children <5 years old averaged ~20 cases per 100,000, but this has dropped dramatically to an average incidence of ~0.15 per 100,000 following licensure of effective vaccines in the late 1980s (Briere et al. 2014). Early passive immunotherapy studies in humans indicated antibodies could play an important role in controlling disease (Beck and Janney 1947), and these results were confirmed in later studies of at-risk children prophylactically treated with hyperimmune globulin (Santosham et al. 1990). Hib anti-capsular antibody was identified as the mechanism of protection as early as 1944 using a passive immunity mouse protection model (Alexander et al. 1944) and these studies were cited as a key motivation for the initial development and clinical testing of several capsular polysaccharide vaccine candidates (Schneerson et al. 1971; Anderson et al. 1972). This history demonstrates that, as with tetanus and diphtheria, vaccine development for Hib benefited from a clear antigen target for antibody-mediated protection and was an approach supported by passive immunity proof-of-principle studies carried out in both animals and humans.

The first Hib polysaccharide vaccine was licensed for use in the US in 1985 (Briere et al. 2014). Pre-licensure studies had estimated VE of 90% (95% CI 58–99%) in 18–71 month old children, but also demonstrated low antibody responses and no efficacy in children <18 months old (Peltola et al. 1984; Ward et al. 1988). Post-licensure evaluation through several case-control studies suggested more modest VE of only 50% in children ≥ 18 months of age (Ward et al. 1988). Immunogenicity was greatly enhanced with the development of polysaccharide-protein conjugate vaccines, with several formulations licensed for use in children as young as 2 months old by 1990 (Briere et al. 2014). Numerous controlled trials estimated the efficacy of the conjugate vaccines at ~90% (Heath 1998), results which have been substantiated by the dramatic decrease of invasive Hib across multiple countries following widespread vaccination (Briere et al. 2014; Heath 1998). Current US recommendations suggest children should receive a 2–3 dose primary vaccination series (depending on the manufacturer) with a final booster dose typically given between 12–15 months of age (Briere et al. 2014). Following a similar vaccination schedule in UK infants, 98% of children were found to maintain Hib capsular polysaccharide antibody titers above the putative protective threshold (defined as 0.15 μg/mL (Anderson 1984)) for up to 2 years following the final booster vaccination (Borrow et al. 2010). A similar study in Spain indicated no drop in geometric mean antibody concentrations from 4.5 to 5.5 years following the final booster vaccination, with >97% of children maintaining antibody concentrations above the 0.15 μg/mL threshold (Tejedor et al. 2012). In total, these results indicate that the Hib conjugate vaccine can induce long-term antibody responses and effectively control this invasive and life-threatening disease.

4.3. Bordetella pertussis

Bordetella pertussis, the bacterial pathogen responsible for the disease commonly known as whooping cough, was at one time a leading cause of childhood morbidity and mortality in the US (Morse 1913). In the decade just prior to vaccine licensure (1932–1941), annual reported attack rates averaged 157 per 100,000 persons, though the true rate has been estimated to be as high as 872 per 100,000 (Cherry 1999). Following the implementation of a whole-cell inactivated vaccine in the 1940s, disease burden dropped dramatically to a low of 0.5 cases per 100,000 persons by 1976 (Faulkner et al. 2015). While the positive public health impact of the whole-cell pertussis (wP) vaccine was striking, with estimates of VE ranging from 70–90% (Advisory Committee on Immunization Practices 1997), the vaccine elicited a relatively high rate of local and systemic adverse events (Cody et al. 1981). The most concerning adverse event was a potential link between vaccination and permanent brain damage due to encephalopathy (Walton et al. 2015; Shorvon and Berg 2008; Allen 2013). However, several of the reported case studies supporting this hypothesis have since been rediagnosed as Dravet syndrome, a form of epilepsy associated with specific genetic mutations (Reyes et al. 2011; Berkovic et al. 2006), and reappraisal of earlier studies calls into question whether any link between wP vaccination and encephalopathy truly exists (Shorvon and Berg 2008; Allen 2013).

Nevertheless, given the public concern surrounding the safety of the wP vaccine, several replacement subunit aP candidates were developed. Unlike tetanus, diphtheria, and Hib, one of the major limitations with the development of subunit vaccines for pertussis was the lack of a defined correlate of immunity against a specific B. pertussis component for vaccine-induced immunity. Early studies indicated a strong correlation between high serum agglutinin titers and protection from disease (Sako 1947; Medical Research Council 1956), but agglutinogens have been reported to include multiple targets such as pertactin (PRN), fimbriae-2 and 3 (FIM-2 and FIM-3), and lipooligosaccharide (LOS) (Cherry et al. 1998). Passive immunity studies performed in a mouse aerosol challenge model demonstrated that monoclonal antibodies against the pertussis toxin (PT), filamentous hemagglutinin (FHA) and FIM-2 could each protect mice against disease (Sato and Sato 1990), underscoring the complexity of the problem. Clinical results from early efficacy trials of aP vaccine candidates were likewise complex, suggesting that combinations of pre-existing antibody titers to a range of antigens, including PT, PRN and FIM-2, could be correlated with vaccine efficacy (Cherry et al. 1998; Storsaeter et al. 1998), but antibodies to FHA appeared to play no role in protection (Cherry et al. 1998), and no single measure seemed to assure protection.

Despite an uncertain understanding of the correlates of immunity associated with aP vaccines, several formulations were licensed for use in the US by 1991 (Advisory Committee on Immunization Practices 1997) and contain a range of proteins including FHA, PRN, FIM-2, FIM-3, and inactivated pertussis toxoid. Current US recommendations for children include a 5-dose schedule of DTaP (diphtheria, tetanus, acellular pertussis) through 4–6 years of age, with a Tdap booster (containing reduced content of both the diphtheria and pertussis components) at 11–12 years of age (Faulkner et al. 2015). Initial estimates of vaccine efficacy appeared promising and ranged from 84–89%, and use of the aP vaccine was recommended for all vaccinations by 1997 (Advisory Committee on Immunization Practices 1997), with the wP vaccine phased out soon thereafter. However, in the decades following implementation of the full aP vaccine schedule, an alarming rise in pertussis cases was observed in several geographical regions including North America, Australia and Europe (Plotkin 2014). Based on the close proximity in time between the resurgence of pertussis and the switch to the aP formulation, Australian researchers examined the relative risk for children developing pertussis based on their immunization history with either the wP or aP vaccines (Sheridan et al. 2012). Their study found that children who received a full aP vaccine schedule had a 3.29-fold increase (95% CI 2.44–4.46) in annual reporting rates for pertussis versus their wP-vaccinated counterparts. Furthermore, in mixed vaccination series (i.e., children receiving both wP and aP formulations) they found that the order of the vaccination series had a substantial impact on outcome, with aP primo-vaccinated children still demonstrating deficits in protection even when given the wP vaccine in later booster doses. This study has been supported by evidence from several US cohorts (Liko et al. 2013; Klein et al. 2013). In Oregon, analysis of children immunized during the transition from DTwP to DTaP demonstrated that priming with the acellular vaccine consistently increased the risk of contracting pertussis by ~2-fold across multiple vaccine booster scenarios (Liko et al. 2013). A case-control study performed in California indicated that teenagers who had received four doses of DTaP versus DTwP formulations during their initial vaccination series had a nearly 6-fold increased risk of contracting PCR-positive pertussis (Klein et al. 2013).

While the underlying cause of poor protective efficacy is unclear, waning immunity may play a key role. A large-scale assessment of the 5-dose DTaP schedule used in the US estimated VE as a function of time (Misegades et al. 2012). This study found VE ranging from 92.3–98.1% during the first 3 years following the final booster dose, but this decreased over time to as low as 71.2% at time points >5 years post-vaccination. In 2006, the Advisory Committee on Immunization Practices (ACIP) recommended adding a Tdap booster at 11–12 years of age in an attempt to counteract this waning immunity (Faulkner et al. 2015). However, boosting immunity in adolescents again appears to be limited. In California, researchers estimated VE of only 68.8% (95% CI 59.7 to 75.9) during the first year after Tdap vaccination of adolescents, dropping to as low as 8.9% (95% CI – 30.6 to 36.4) by the fourth year (Klein et al. 2016). Studies in both Wisconsin (Koepke et al. 2014) and Washington (Acosta et al. 2015) substantiate these results, with VE ranging from 73.3%−75% during the first year following vaccination, but rapidly waning to 11.9%−34% within just a few years. The answer to this public health problem is unclear. A range of potential solutions have been proposed and although a return to the old wP vaccine is considered unlikely (Plotkin 2014), new wP vaccines with high efficacy and improved safety profiles might still be a viable option. Increasing antigen doses may improve immunogenicity since VE differences have been observed between different Tdap manufacturers, with higher antigen doses associated with increased VE (Koepke et al. 2014). However even with increased antigen dose, VE still wanes rapidly, pointing to what may be a fundamental limitation of the current acellular vaccine approach. Until researchers can find an improved approach to elicit durable immunity while retaining an acceptable safety profile, pertussis will represent a continuing problem for the foreseeable future.

5. Partially successful vaccines

In addition to examples of successful vaccines and clear vaccine failures, there are also a large number of partially successful vaccines. We define a vaccine as “partially successful” if it elicits a protective immune response against a particular pathogen with efficacy rates that are near or substantially less than 50%. Challenging pathogens, such as those with multiple serotypes, complex lifecycles, or high antigenic variability are the ones that are most likely to have vaccine counterparts that are either non-existent or only partially successful. Some examples of challenging pathogens with partially successful vaccine candidates include (a) dengue virus (DENV) with its 4 inter-related serotypes, (b) VZV representing a latent herpesvirus that causes chickenpox (primary infection) or shingles (after reactivation/re-exposure), (c) Plasmodium, a parasite with a complex multi-stage lifecycle that causes malaria, and (d) human immunodeficiency virus (HIV), a retrovirus with a complex lifecycle as well as high antigenic variability.

5.1. Dengue virus

DENV is a mosquito-borne pathogen that is ubiquitous throughout tropical regions of the world and endemic or epidemic in 154 countries (Furuya-Kanamori et al. 2016). DENV infection may result in a spectrum of disease ranging from acute, debilitating febrile illness (dengue fever, DF) to severe, life-threatening hemorrhagic disease (dengue hemorrhagic fever, DHF; dengue shock syndrome, DSS). Within DENV, there are 4 serotypes (DENV1, DENV2, DENV3, and DENV4), and each has been found to cause human disease and mortality. Current estimates indicate that nearly 4 billion people are at risk (Brady et al. 2012), with disease burden ranging from 58–96 million apparent DENV infections per year (Bhatt et al. 2013; Stanaway et al. 2016), resulting in approximately 500,000 cases of DHF/DSS and greater than 20,000 deaths (Murray et al. 2013). Infection with one DENV serotype is known to provide durable protective immunity against reinfection by that particular serotype (Webster et al. 2009). However, immunity to one DENV serotype may predispose an individual to more severe disease if infected by a second DENV serotype through a process believed to be associated with antibody-dependent enhancement (ADE) of infection of Fc-γ receptor-bearing cells (Guzman et al. 2000; Burke et al. 1988; Vaughn et al. 2000; Guzman et al. 1987; Halstead et al. 1970). Although the role of ADE in DHF/DSS has been questioned in recent years (Webster et al. 2009), this concept nonetheless remains an important consideration in terms of vaccine development. As such, it is widely accepted that an optimal DENV vaccine must provide protection against all 4 serotypes to provide broad antiviral immunity and to prevent the potential complications of ADE.

Dengvaxia® is a live, attenuated tetravalent vaccine that consists of four chimeric yellow fever virus strain 17D (YFV-17D) vectors expressing the DENV envelope and premembrane proteins of each DENV serotype, and has recently become the first licensed DENV vaccine (Sanofi Pasteur 2016). Despite the excitement surrounding this major milestone, concerns have emerged over efficacy and safety (Simmons 2015; Halstead and Russell 2016). An initial Phase IIb trial conducted in Thailand estimated an overall VE of only 30.2% (95% CI - 13.4– 56.6) and the primary estimate of efficacy was not significant (Sabchareon et al. 2012). When segregated by serotype, there was some indication of efficacy for DENV1, DENV3 and DENV4, whereas with DENV2 (which constituted the majority of cases among vaccinated and controls) the vaccine demonstrated no protection. Larger Phase III trials in both Asia (Capeding et al. 2014) and Latin America (Villar et al. 2015) have now demonstrated modest protection against DENV, with VE ranging from 56.5–60.8%. However, as suggested in the Phase IIb trial, both studies demonstrated differences among serotypes, with DENV2 again showing the lowest protective immunity (35.0–42.3% VE). More concerning may be the limited protection observed in younger subjects, and those who were seronegative at the time of vaccination. In the Asian trial, participant age ranged from 2–14 years old, and decreasing efficacy was closely tied to decreasing age, with 2–5 year old children demonstrating only 33.7% VE (95% CI 11.7–50.0) (Capeding et al. 2014). The Latin American trial only recruited children 9–16 years of age, and found similar efficacy across this range (Villar et al. 2015). Across both Phase III trials, while VE in seropositive children ranged from 74.3–83.7%, it was sharply lower among previously seronegative children (VE = 35.5–43.2%) (Capeding et al. 2014; Villar et al. 2015). This suggests the age dependence observed in the Asian trial may be tied to the rates of seropositivity prior to vaccination within age groups (i.e., younger children have a lower likelihood of prior exposure to wild type DENV). Seronegative children demonstrated neutralizing antibody titers that were typically ~10-fold lower than their seropositive counterparts after vaccination (Table S8 in (Villar et al. 2015)), and this limited immune response could play a role in the low observed VE. Unfortunately, this still leaves unexplained why seronegative children were unable to mount a robust immune response even after 3 immunizations spaced every 6 months. These results likely underlie the current recommendation to immunize only children ≥9 years of age (Sanofi Pasteur 2016). Unfortunately, this leaves a substantial gap in coverage, especially considering that younger children represent a vulnerable population that experience the highest rates of severe complications following DENV exposure (Guzman et al. 2002).

Formal estimates of Dengvaxia VE have been limited to one year following the completion of the vaccination series. Ongoing long-term safety analyses have been described (Hadinegoro et al. 2015) and will monitor hospitalization rates for DENV illness in vaccinees and controls for up to 6 years. Through 3–4 years post-vaccination (depending on the trial), there appears to be a retained benefit for children vaccinated at 9–16 years of age (Hadinegoro et al. 2015). Conversely, there may also be a risk of increased hospitalizations for younger subjects, especially those who were 2–5 years old at the time of vaccination. Whether or not this points to enhanced disease among children with suboptimal immune responses remains uncertain, though it has been noted that the frequency of severe forms of DENV (such as DSS) do not appear to be increased in this age group (Simmons 2015). Nevertheless, given the relatively modest efficacy of the vaccine and substantial age restrictions, DENV continues to pose a global threat with limited solutions.

5.2. Varicella zoster virus

Following the initial 82–86% success rate of the live attenuated VZV vaccine for prevention of varicella (i.e., chickenpox) in children after a single dose (Izurieta et al. 1997; Vazquez et al. 2001; Lopez et al. 2006), it was assumed that a similar approach would be successful for protection of older individuals (≥60 years of age) against herpes zoster (i.e., shingles). However, in a pivotal randomized double-blinded clinical trial involving 38,546 adults monitored for a median of 3.12 years, the Oka/Merck VZV vaccine provided 51.3% efficacy against clinical diagnosis of herpes zoster (Oxman et al. 2005). Vaccine efficacy was higher among younger subjects (63.9%) but reached only 37.6% efficacy in subjects 70 years of age or older – indicating that the vaccine performed poorly in the aged population at most risk for developing severe herpes zoster and post-herpetic neuralgia. In 2006, the ACIP recommended a single dose of vaccine against herpes zoster (Harpaz et al. 2008) but in 2013, the ACIP declined to recommend the vaccine for younger adults 50–59 years of age due to the limited amount of information regarding the durability of protection after vaccination (Tseng et al. 2016). Indeed, several studies found evidence of rapidly declining protection (Morrison et al. 2015; Schmader et al. 2012; Tseng et al. 2016) with the most compelling evidence presented in a study comparing 176,078 vaccinated subjects to 528,234 unvaccinated subjects who were followed for up to 8 years after vaccination (Tseng et al. 2016). Although vaccine efficacy approached 69% during the first year after vaccination, protective immunity declined rapidly thereafter with just 4% vaccine efficacy by the 8th year after immunization. The decline in protection against disease was similar among vaccine recipients aged 60–69 years and those aged ≥70 years, indicating that advanced age itself was not responsible for the decline in protective immunity.

Although no proven correlate of protective immunity exists for herpes zoster, a long-held belief has been that loss of antiviral T cell-mediated immunity is the determining risk factor for disease onset (Kost and Straus 1996; Wilson et al. 1992; Miller 1980). Along these lines, studies demonstrated that VZV vaccination resulted in a substantial increase in T cell-mediated immunity (Oxman 1995; Levin et al. 2003) and the dose of vaccine for prevention of herpes zoster (approximately 14-fold higher than the dose administered during primary vaccination to prevent varicella) was chosen on the basis of its ability to elicit a significant increase in antiviral T cell immunity among older adults (Oxman et al. 2005). Bearing in mind that the first-line approach of using a live attenuated strain of VZV for vaccination would be the most likely candidate for eliciting strong antiviral T cell responses, one would have found little support for the use of a non-replicating subunit vaccine since it would presumably be far less likely to elicit the T cell responses that develop following infection with a live attenuated version of the same virus. Nevertheless, conventional wisdom was displaced by the success of a non-traditional approach to vaccination against herpes zoster with a subunit vaccine consisting of a single viral protein antigen, recombinant VZV glycoprotein E (gE) (Lal et al. 2015). In this study involving 15,411 subjects (7,698 vaccinated and 7,713 placebo controls) who were followed for a mean of 3.2 years, vaccine efficacy was 97.2%. There was also no age-associated reduction in vaccine efficacy among subjects ≥70 years of age. This indicated that the immunosenescence and age-associated reduction in vaccine efficacy observed after live VZV vaccination could be overcome with a simple 2-dose regimen of a non-replicating subunit vaccine. Further analysis of the subunit vaccine-induced VZV gE-specific CD4+ T cell responses and gE-specific antibody responses indicates that immunity initially declines for the first 1–2 years after vaccination but then reaches a plateau that is maintained for at least 6 years. In these studies, CD4+ T cell memory was sustained at nearly 4-fold higher than pre-vaccination levels and antiviral antibody responses remain approximately 7-fold higher than pre-vaccination levels (Chlibek et al. 2016). Together, these results indicate that this simple subunit vaccine has the potential to provide long-lasting immunity against herpes zoster.

5.3. Plasmodium

Plasmodium parasites are the causative agent of malaria and the complex lifecycle of this parasite has been problematic for the development of an effective vaccine. The human host is infected by sporozoites transmitted by infected mosquitos. The sporozoites travel through the bloodstream and infect hepatocytes where they reproduce as merozoites. The merozoites infect and multiply in new red blood cells that lyse to release more merozoites and initiate the next round of infection. Some merozoites develop into the precursors of male and female gametes, which can infect a feeding mosquito and mature, fuse to form fertilized ookinetes, and develop into sporozoites that travel to the salivary glands where they can be transmitted to the next human host during blood feeding. The invading sporozoites that are transmitted by infected mosquitos represent the best vaccine target since blocking infection at the earliest stage of transmission is likely to provide the highest clinical benefit. The most advanced malaria vaccine candidate is RTS,S, a subunit vaccine based on a Plasmodium circumsporozoite protein (CSP) that is genetically fused to hepatitis B virus surface antigen. The vaccine has been in development for more than 30 years (Kaslow and Biernaux 2015) and has advanced through Phase III clinical trials. Protection against clinical malaria in children (ages 5–17 months) after 3 doses of vaccine is modest; vaccine efficacy reached 45.1% during the first 0–20 months after vaccination but waned rapidly, with only 16.1% vaccine efficacy observed during months 21–32 in young children. The levels of protection are even lower among infants (ages 6–12 weeks) with only 27.0% protective efficacy at 0–20 months with immunity falling to near negligible levels (7.6%) from 21–32 months after vaccination (RTS-S Clinical Trials Partnership 2015). The partial success among children and the weak protective immunity elicited among infants indicates that this subunit vaccine is unable to reliably protect the most vulnerable populations who are at the highest risk for severe cases of malaria and this is compounded by the lack of durable immunity even after a 3-dose regimen.

An alternative approach to malaria vaccination that has gained recent attention utilizes intact gamma-irradiated sporozoites purified from infected mosquitos (Richie et al. 2015; Hoffman et al. 2015). The main advantage of this approach is that vaccination is performed using the whole organism in which many potentially immunogenic proteins are presented in their native conformation. In a human challenge model of Plasmodium infection, the highest administered dose provided 100% protection from parasitemia for 6 immunized subjects (Seder et al. 2013). The main disadvantages to this approach are that direct intravenous injection of the vaccine is required (early studies involving subcutaneous or intradermal vaccination failed (Epstein et al. 2011)) and there are technical limitations to the production, purification, and long-term stability of sporozoites prepared from Plasmodium-infected mosquitos. Another potential concern is that the vaccine may be on the knife’s edge in terms of protective efficacy since a 4-dose vaccination schedule elicited promastigote-specific antibodies that were roughly half of that obtained from a 5-dose schedule and similarly, protection from clinical disease declined from 100% (6/6 subjects) to 60% (6/9 subjects). Since many vaccine-mediated immune responses decline by 2- to 10-fold from their peak levels before reaching a plateau stage of more stable long-term maintenance, it is possible that this vaccine approach may lose at least some protective efficacy over a relatively short period of time. Nevertheless, the results of these studies provide a foundation on which future vaccines may be developed that elicit appropriate immune responses that block the earliest stages of Plasmodium infection and further studies are currently underway (Hoffman et al. 2015).

5.4. Human immunodeficiency virus

HIV, the causative agent of Acquired Immune Deficiency Syndrome (AIDS), is a notoriously difficult vaccine target and of the 6 field efficacy trials that have been performed to date, all HIV vaccines have failed except one, the RV144 vaccine (Kim et al. 2015). The RV144 trial involved 16,402 subjects and a prime-boost strategy utilizing a recombinant canarypox vector expressing HIV Gag, pro, and envelope administered at 0, 1, 3, and 6 months in addition to alum-adjuvanted monomeric envelope proteins co-administered at the 3 and 6 month time points. Vaccine efficacy reached 60.5% at 1 year after vaccination but declined to 31.2% efficacy at 3.5 years after vaccination (Kim et al. 2015). Despite these suboptimal results, any significant protection against HIV is encouraging and current clinical trials involve redesigned gp120 envelope constructs, comparison of different adjuvants (MF59 and ASO1B) and a revised vaccination schedule that includes a booster dose administered at 12 months (Gray et al. 2016). Moreover, development of intact HIV envelope trimers provide a new approach to HIV vaccine development and further studies are planned to test multiple trimers in either simultaneous or sequential combinations in order to optimally induce high titer, broadly neutralizing antibodies (de Taeye et al. 2016). Regardless of the approach that is taken, development of a vaccination strategy that induces long-term immunity above a protective threshold (preferably with broadly neutralizing antibodies (Burton and Mascola 2015)) is a high priority and likely to be key to the success of any future HIV vaccine.

6. Future vaccines for challenging pathogens

Despite the ultimate goal of performing rationale vaccine design in which a deeper understanding of pathogen structure, immunogenicity, and pathogenesis is utilized in the construction of a new vaccine, many of our current successful vaccine strategies have been developed empirically. In some cases such as measles, mumps, and rubella, an attenuated version of the target virus provides safe and effective vaccine-mediated protection. In other cases, successful vaccine development was counterintuitive to the approach that one might have predicted based on the characteristics of the pathogen itself. HPV for example, represents a mucosal pathogen that evades the immune system in order to sustain a chronic, essentially lifelong infection within its human host. One might have predicted that a mucosal route of vaccination would be preferred, that CD8+ T cells would be the most important cell type for protecting against intracellular pathogens, and that antiviral IgA responses might be much more important than IgG responses in terms of protecting mucosal sites from natural infection. In contrast to these a priori expectations, development of non-replicating HPV VLP vaccines that are administered by intramuscular injection induce strong IgG responses and relatively minor IgA or T cell responses but still provide sustained protective immunity against HPV for many years after vaccination. This result would not have been predicted based on conventional wisdom and knowledge of the natural host:pathogen interactions and provides an interesting counterpoint showing that sometimes it is important to take empirical approaches to the development of vaccines to the most complex pathogens.

Acknowledgements:

This work was funded in part with federal funds from the National Institute of Allergy and Infectious Diseases R44 AI079898 (to MKS and IJA), R01 AI098723 (to MKS) and Oregon National Primate Research Center grant, 8P51 OD01109253 (to MKS). OHSU, Dr. Slifka, and Dr. Amanna have a financial interest in Najít Technologies, Inc., a company that is developing a new dengue virus vaccine based on a hydrogen peroxide-based inactivation approach. This potential individual and institutional conflict of interest has been reviewed and managed by OHSU. No writing assistance was utilized in the production of this manuscript.

Contributor Information

Ian J. Amanna, Najít Technologies, Inc., Beaverton, OR 97006, USA.

Mark K. Slifka, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006

References

  1. Acosta AM, DeBolt C, Tasslimi A, Lewis M, Stewart LK, Misegades LK, Messonnier NE, Clark TA, Martin SW, Patel M (2015) Tdap vaccine effectiveness in adolescents during the 2012 Washington State pertussis epidemic. Pediatrics 135:981–989. doi: 10.1542/peds.2014-3358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Acosta PL, Caballero MT, Polack FP (2016) Brief History and Characterization of Enhanced Respiratory Syncytial Virus Disease. Clin Vaccine Immunol 23:189–195. doi: 10.1128/CVI.00609-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Adams D, Fullerton K, Jajosky R, Sharp P, Onweh D, Schley A, Anderson W, Faulkner A, Kugeler K (2015) Summary of Notifiable Infectious Diseases and Conditions - United States, 2013. MMWR Morb Mortal Wkly Rep 62 (53):1–122. doi: 10.15585/mmwr.mm6253a1 [DOI] [PubMed] [Google Scholar]
  4. Advisory Committee on Immunization Practices (1997) Pertussis Vaccination: Use of Acellular Pertussis Vaccines Among Infants and Young Children. MMWR Recomm Rep 46 (RR-7):1–32 [PubMed] [Google Scholar]
  5. Advisory Committee on Immunization Practices (2000) Poliomyelitis prevention in the United States. Updated recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 49 (RR-5):1–22 [PubMed] [Google Scholar]
  6. Alexander HE, Heidelberger M, Leidy G (1944) The Protective or Curative Element in Type B H. influenzae Rabbit Serum. Yale J Biol Med 16 (5):425–434 [PMC free article] [PubMed] [Google Scholar]
  7. Alexander LN, Seward JF, Santibanez TA, Pallansch MA, Kew OM, Prevots DR, Strebel PM, Cono J, Wharton M, Orenstein WA, Sutter RW (2004) Vaccine policy changes and epidemiology of poliomyelitis in the United States. Jama 292 (14):1696–1701 [DOI] [PubMed] [Google Scholar]
  8. Allen A (2013) Public health. The pertussis paradox. Science 341:454–455. doi: 10.1126/science.341.6145.454 [DOI] [PubMed] [Google Scholar]
  9. Amanna IJ, Carlson NE, Slifka MK (2007) Duration of humoral immunity to common viral and vaccine antigens. N Engl J Med 357 (19):1903–1915 [DOI] [PubMed] [Google Scholar]
  10. Amanna IJ, Messaoudi I, Slifka MK (2008) Protective immunity following vaccination: how is it defined? Hum Vaccin 4 (4):316–319. doi:5751 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Amanna IJ, Slifka MK (2010) Mechanisms that determine plasma cell lifespan and the duration of humoral immunity. Immunol Rev 236:125–138. doi:IMR912 [pii] 10.1111/j.1600-065X.2010.00912.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Anderson P (1984) The protective level of serum antibodies to the capsular polysaccharide of Haemophilus influenzae type b. J Infect Dis 149 (6):1034–1035 [DOI] [PubMed] [Google Scholar]
  13. Anderson P, Johnston RB Jr., Smith DH (1972) Human serum activities against Hemophilus influenzae, type b. J Clin Invest 51 (1):31–38. doi: 10.1172/JCI106793 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Beck KH, Janney FR (1947) Alexander’s rabbit serum in the treatment of influenzal meningitis; an evaluation of its use in conjunction with sulfonamide compounds. JAMA 73 (3):317–325 [DOI] [PubMed] [Google Scholar]
  15. Beem M, Wright FH, Hamre D, Egerer R, Oehme M (1960) Association of the chimpanzee coryza agent with acute respiratory disease in children. N Engl J Med 263:523–530. doi: 10.1056/NEJM196009152631101 [DOI] [PubMed] [Google Scholar]
  16. Berkovic SF, Harkin L, McMahon JM, Pelekanos JT, Zuberi SM, Wirrell EC, Gill DS, Iona X, Mulley JC, Scheffer IE (2006) De-novo mutations of the sodium channel gene SCN1A in alleged vaccine encephalopathy: a retrospective study. Lancet Neurol 5:488–492. doi: 10.1016/S1474-4422(06)70446-X [DOI] [PubMed] [Google Scholar]
  17. Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, Drake JM, Brownstein JS, Hoen AG, Sankoh O, Myers MF, George DB, Jaenisch T, Wint GR, Simmons CP, Scott TW, Farrar JJ, Hay SI (2013) The global distribution and burden of dengue. Nature 496:504–507. doi: 10.1038/nature12060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bjorkholm B, Bottiger M, Christenson B, Hagberg L (1986) Antitoxin antibody levels and the outcome of illness during an outbreak of diphtheria among alcoholics. Scandinavian journal of infectious diseases 18 (3):235–239 [DOI] [PubMed] [Google Scholar]
  19. Black S, Friedland LR, Schuind A, Howe B (2006) Immunogenicity and safety of a combined DTaP-IPV vaccine compared with separate DTaP and IPV vaccines when administered as pre-school booster doses with a second dose of MMR vaccine to healthy children aged 4–6 years. Vaccine 24:6163–6171. doi: 10.1016/j.vaccine.2006.04.001 [DOI] [PubMed] [Google Scholar]
  20. Bonsignori M, Moody MA, Parks RJ, Holl TM, Kelsoe G, Hicks CB, Vandergrift N, Tomaras GD, Haynes BF (2009) HIV-1 envelope induces memory B cell responses that correlate with plasma antibody levels after envelope gp120 protein vaccination or HIV-1 infection. J Immunol 183:2708–2717. doi: 10.4049/jimmunol.0901068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Borrow R, Andrews N, Findlow H, Waight P, Southern J, Crowley-Luke A, Stapley L, England A, Findlow J, Miller E (2010) Kinetics of antibody persistence following administration of a combination meningococcal serogroup C and haemophilus influenzae type b conjugate vaccine in healthy infants in the United Kingdom primed with a monovalent meningococcal serogroup C vaccine. Clin Vaccine Immunol 17:154–159. doi: 10.1128/CVI.00384-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Brady OJ, Gething PW, Bhatt S, Messina JP, Brownstein JS, Hoen AG, Moyes CL, Farlow AW, Scott TW, Hay SI (2012) Refining the global spatial limits of dengue virus transmission by evidence-based consensus. PLoS Negl Trop Dis 6:e1760. doi: 10.1371/journal.pntd.0001760 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Briere EC, Rubin L, Moro PL, Cohn A, Clark T, Messonnier N (2014) Prevention and control of haemophilus influenzae type b disease: recommendations of the advisory committee on immunization practices (ACIP). MMWR Recomm Rep 63:1–14 [PubMed] [Google Scholar]
  24. Burke DS, Nisalak A, Johnson DE, Scott RM (1988) A prospective study of dengue infections in Bangkok. The American journal of tropical medicine and hygiene 38 (1):172–180 [DOI] [PubMed] [Google Scholar]
  25. Burton DR, Mascola JR (2015) Antibody responses to envelope glycoproteins in HIV-1 infection. Nat Immunol 16 (6):571–576. doi: 10.1038/ni.3158 ni.3158 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Capeding MR, Tran NH, Hadinegoro SR, Ismail HI, Chotpitayasunondh T, Chua MN, Luong CQ, Rusmil K, Wirawan DN, Nallusamy R, Pitisuttithum P, Thisyakorn U, Yoon IK, van der Vliet D, Langevin E, Laot T, Hutagalung Y, Frago C, Boaz M, Wartel TA, Tornieporth NG, Saville M, Bouckenooghe A, Group CYDS (2014) Clinical efficacy and safety of a novel tetravalent dengue vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial. Lancet 384 (9951):1358–1365. doi: 10.1016/S0140-6736(14)61060-6 [DOI] [PubMed] [Google Scholar]
  27. Carter CH, Conway TJ, Cornfeld D, Iezzoni DG, Kempe CH, Moscovici C, Rauh LW, Vignec AJ, Warren J (1962) Serologic response of children to inactivated measles vaccine. JAMA 179:848–853 [DOI] [PubMed] [Google Scholar]
  28. Centers for Disease Control and Prevention (1996) Prevention of hepatitis A through active or passive immunization: Recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 45 (RR-15):1–30 [PubMed] [Google Scholar]
  29. Centers for Disease Control and Prevention (2015a) Cervical Cancer Statistics. Centers for Disease Control and Prevention,. http://www.cdc.gov/cancer/cervical/statistics/-2. Accessed April 18 2016
  30. Centers for Disease Control and Prevention (2015b) Diphtheria In: Hamborsky J, Kroger A, Wolfe S (eds) Epidemiology and prevention of vaccine-preventable diseases. 13th edn Public Health Foundation, Washington DC, pp 107–118 [Google Scholar]
  31. Centers for Disease Control and Prevention (2015c) Hepatitis A In: Hamborsky J, Kroger A, Wolfe S (eds) Epidemiology and prevention of vaccine-preventable diseases. 13th edn Public Health Foundation, Washington DC, pp 135–148 [Google Scholar]
  32. Centers for Disease Control and Prevention (2015d) Human papillomavirus In: Hamborsky J, Kroger A, Wolfe S (eds) Epidemiology and prevention of vaccine-preventable diseases. 13th edn Public Health Foundation, Washington DC, pp 175–186 [Google Scholar]
  33. Centers for Disease Control and Prevention (2015e) Measles In: Hamborsky J, Kroger A, Wolfe S (eds) Epidemiology and prevention of vaccine-preventable diseases. 13th edn Public Health Foundation, Washington DC, pp 209–230 [Google Scholar]
  34. Centers for Disease Control and Prevention (2015f) Tetanus In: Hamborsky J, Kroger A, Wolfe S (eds) Epidemiology and prevention of vaccine-preventable diseases. 13th edn Public Health Foundation, Washington DC, pp 341–352 [Google Scholar]
  35. Chanock RM, Kim HW, Vargosko AJ, Deleva A, Johnson KM, Cumming C, Parrott RH (1961) Respiratory syncytial virus. I. Virus recovery and other observations during 1960 outbreak of bronchiolitis, pneumonia, and minor respiratory diseases in children. JAMA 176:647–653 [PubMed] [Google Scholar]
  36. Cherry JD (1999) Pertussis in the preantibiotic and prevaccine era, with emphasis on adult pertussis. Clin Infect Dis 28 Suppl 2:S107–111. doi: 10.1086/515057 [DOI] [PubMed] [Google Scholar]
  37. Cherry JD, Gornbein J, Heininger U, Stehr K (1998) A search for serologic correlates of immunity to Bordetella pertussis cough illnesses. Vaccine 16:1901–1906 [DOI] [PubMed] [Google Scholar]
  38. Chin J, Magoffin RL, Shearer LA, Schieble JH, Lennette EH (1969) Field evaluation of a respiratory syncytial virus vaccine and a trivalent parainfluenza virus vaccine in a pediatric population. Am J Epidemiol 89 (4):449–463 [DOI] [PubMed] [Google Scholar]
  39. Chlibek R, Pauksens K, Rombo L, van Rijckevorsel G, Richardus JH, Plassmann G, Schwarz TF, Catteau G, Lal H, Heineman TC (2016) Long-term immunogenicity and safety of an investigational herpes zoster subunit vaccine in older adults. Vaccine 34 (6):863–868. doi: 10.1016/j.vaccine.2015.09.073 S0264-410X(15)01349-3 [pii] [DOI] [PubMed] [Google Scholar]
  40. Cody CL, Baraff LJ, Cherry JD, Marcy SM, Manclark CR (1981) Nature and rates of adverse reactions associated with DTP and DT immunizations in infants and children. Pediatrics 68 (5):650–660 [PubMed] [Google Scholar]
  41. Day PM, Kines RC, Thompson CD, Jagu S, Roden RB, Lowy DR, Schiller JT (2010) In vivo mechanisms of vaccine-induced protection against HPV infection. Cell Host Microbe 8:260–270. doi: 10.1016/j.chom.2010.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. de Taeye SW, Moore JP, Sanders RW (2016) HIV-1 Envelope Trimer Design and Immunization Strategies To Induce Broadly Neutralizing Antibodies. Trends Immunol 37 (3):221–232. doi: 10.1016/j.it.2016.01.007 S1471-4906(16)00008-9 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Einstein MH, Levin MJ, Chatterjee A, Chakhtoura N, Takacs P, Catteau G, Dessy FJ, Moris P, Lin L, Struyf F, Dubin G, Group HPVS (2014) Comparative humoral and cellular immunogenicity and safety of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine and HPV-6/11/16/18 vaccine in healthy women aged 18–45 years: follow-up through Month 48 in a Phase III randomized study. Hum Vaccin Immunother 10 (12):3455–3465. doi: 10.4161/hv.36117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Embree J, Law B, Voloshen T, Tomovici A (2015) Immunogenicity, safety, and antibody persistence at 3, 5, and 10 years postvaccination in adolescents randomized to booster immunization with a combined tetanus, diphtheria, 5-component acellular pertussis, and inactivated poliomyelitis vaccine administered with a hepatitis B virus vaccine concurrently or 1 month apart. Clin Vaccine Immunol 22:282–290. doi: 10.1128/CVI.00682-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Epstein JE, Tewari K, Lyke KE, Sim BK, Billingsley PF, Laurens MB, Gunasekera A, Chakravarty S, James ER, Sedegah M, Richman A, Velmurugan S, Reyes S, Li M, Tucker K, Ahumada A, Ruben AJ, Li T, Stafford R, Eappen AG, Tamminga C, Bennett JW, Ockenhouse CF, Murphy JR, Komisar J, Thomas N, Loyevsky M, Birkett A, Plowe CV, Loucq C, Edelman R, Richie TL, Seder RA, Hoffman SL (2011) Live attenuated malaria vaccine designed to protect through hepatic CD8(+) T cell immunity. Science 334 (6055):475–480. doi: 10.1126/science.1211548 science.1211548 [pii] [DOI] [PubMed] [Google Scholar]
  46. Faulkner A, Skoff T, Martin S, Cassiday P, Tondella ML, Liang J (2015) Chapter 10: Pertussis. In: Manual for the surveillance of vaccine-preventable diseases. Centers for Disease Control and Prevention, Atlanta, GA, [Google Scholar]
  47. Feltes TF, Cabalka AK, Meissner HC, Piazza FM, Carlin DA, Top FH Jr., Connor EM, Sondheimer HM (2003) Palivizumab prophylaxis reduces hospitalization due to respiratory syncytial virus in young children with hemodynamically significant congenital heart disease. J Pediatr 143:532–540 [DOI] [PubMed] [Google Scholar]
  48. Fulginiti VA, Eller JJ, Downie AW, Kempe CH (1967) Altered reactivity to measles virus. Atypical measles in children previously immunized with inactivated measles virus vaccines. JAMA 202 (12):1075–1080 [DOI] [PubMed] [Google Scholar]
  49. Fulginiti VA, Eller JJ, Sieber OF, Joyner JW, Minamitani M, Meiklejohn G (1969) Respiratory virus immunization. I. A field trial of two inactivated respiratory virus vaccines; an aqueous trivalent parainfluenza virus vaccine and an alum-precipitated respiratory syncytial virus vaccine. Am J Epidemiol 89 (4):435–448 [DOI] [PubMed] [Google Scholar]
  50. Furuya-Kanamori L, Liang S, Milinovich G, Soares Magalhaes RJ, Clements AC, Hu W, Brasil P, Frentiu FD, Dunning R, Yakob L (2016) Co-distribution and co-infection of chikungunya and dengue viruses. BMC Infect Dis 16:84. doi: 10.1186/s12879-016-1417-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Gellis SS, Stokes J Jr., Brother GM, Hall WM, Gilmore HR, Beyer E, Morrissey RA (1945) The use of human immune serum globulin (gamma globulin) in infectious (epidemic) hepatitis in the Mediterranean theater of operations I. Studies on prophylaxis in two epidemics of infectious hepatitis. JAMA 128 (15):1062–1063 [Google Scholar]
  52. Giannini SL, Hanon E, Moris P, Van Mechelen M, Morel S, Dessy F, Fourneau MA, Colau B, Suzich J, Losonksy G, Martin MT, Dubin G, Wettendorff MA (2006) Enhanced humoral and memory B cellular immunity using HPV16/18 L1 VLP vaccine formulated with the MPL/aluminium salt combination (AS04) compared to aluminium salt only. Vaccine 24 (33–34):5937–5949. doi:S0264-410X(06)00709-2 [pii] 10.1016/j.vaccine.2006.06.005 [DOI] [PubMed] [Google Scholar]
  53. Glenn GM, Fries LF, Thomas DN, Smith G, Kpamegan E, Lu H, Flyer D, Jani D, Hickman SP, Piedra PA (2016) A Randomized, Blinded, Controlled, Dose-Ranging Study of a Respiratory Syncytial Virus Recombinant Fusion (F) Nanoparticle Vaccine in Healthy Women of Childbearing Age. J Infect Dis 213:411–422. doi: 10.1093/infdis/jiv406 [DOI] [PubMed] [Google Scholar]
  54. Goulon M, Girard O, Grosbuis S, Desormeau JP, Capponi MF (1972) [Antitetanus antibodies. Assay before anatoxinotherapy in 64 tetanus patients]. Nouv Presse Med 1 (45):3049–3050 [PubMed] [Google Scholar]
  55. Gray GE, Laher F, Lazarus E, Ensoli B, Corey L (2016) Approaches to preventative and therapeutic HIV vaccines. Curr Opin Virol 17:104–109. doi:S1879-6257(16)30014-1 [pii] 10.1016/j.coviro.2016.02.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Guinee VF, Henderson DA, Casey HL, Wingo ST, Ruthig DW, Cockburn TA, Vinson TO, Calafiore DC, Winkelstein W Jr., Karzon DT, Rathbun ML, Alexander ER, Peterson DR (1966) Cooperative measles vaccine field trial. I. Clinical efficacy. Pediatrics 37 (4):649–665 [PubMed] [Google Scholar]
  57. Guzman MG, Kouri G, Bravo J, Valdes L, Vazquez S, Halstead SB (2002) Effect of age on outcome of secondary dengue 2 infections. Int J Infect Dis 6 (2):118–124 [DOI] [PubMed] [Google Scholar]
  58. Guzman MG, Kouri G, Martinez E, Bravo J, Riveron R, Soler M, Vazquez S, Morier L (1987) Clinical and serologic study of Cuban children with dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). Bull Pan Am Health Organ 21 (3):270–279 [PubMed] [Google Scholar]
  59. Guzman MG, Kouri G, Valdes L, Bravo J, Alvarez M, Vazques S, Delgado I, Halstead SB (2000) Epidemiologic studies on Dengue in Santiago de Cuba, 1997. Am J Epidemiol 152 (9):793–799; discussion 804 [DOI] [PubMed] [Google Scholar]
  60. Hadinegoro SR, Arredondo-Garcia JL, Capeding MR, Deseda C, Chotpitayasunondh T, Dietze R, Muhammad Ismail HI, Reynales H, Limkittikul K, Rivera-Medina DM, Tran HN, Bouckenooghe A, Chansinghakul D, Cortes M, Fanouillere K, Forrat R, Frago C, Gailhardou S, Jackson N, Noriega F, Plennevaux E, Wartel TA, Zambrano B, Saville M, Group C-TDVW (2015) Efficacy and Long-Term Safety of a Dengue Vaccine in Regions of Endemic Disease. N Engl J Med 373 (13):1195–1206. doi: 10.1056/NEJMoa1506223 [DOI] [PubMed] [Google Scholar]
  61. Hall CB, Weinberg GA, Iwane MK, Blumkin AK, Edwards KM, Staat MA, Auinger P, Griffin MR, Poehling KA, Erdman D, Grijalva CG, Zhu Y, Szilagyi P (2009) The burden of respiratory syncytial virus infection in young children. N Engl J Med 360:588–598. doi: 10.1056/NEJMoa0804877 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Halstead SB, Nimmannitya S, Cohen SN (1970) Observations related to pathogenesis of dengue hemorrhagic fever. IV. Relation of disease severity to antibody response and virus recovered. Yale J Biol Med 42 (5):311–328 [PMC free article] [PubMed] [Google Scholar]
  63. Halstead SB, Russell PK (2016) Protective and immunological behavior of chimeric yellow fever dengue vaccine. Vaccine 34 (14):1643–1647. doi: 10.1016/j.vaccine.2016.02.004 [DOI] [PubMed] [Google Scholar]
  64. Hammarlund E, Thomas A, Poore EA, Amanna IJ, Rynko AE, Mori M, Chen Z, Slifka MK (2016) Durability of Vaccine-Induced Immunity Against Tetanus and Diphtheria Toxins: A Cross-sectional Analysis. Clin Infect Dis 62:1111–1118. doi: 10.1093/cid/ciw066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hao L, Toyokawa S, Kobayashi Y (2008) Poisson-model analysis of the risk of vaccine-associated paralytic poliomyelitis in Japan between 1971 and 2000. Jpn J Infect Dis 61 (2):100–103 [PubMed] [Google Scholar]
  66. Harpaz R, Ortega-Sanchez IR, Seward JF (2008) Prevention of herpes zoster: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 57 (RR-5):1–30; quiz CE32–34. doi:rr5705a1 [pii] [PubMed] [Google Scholar]
  67. Havens WP Jr., Paul JR (1945) Prevention of infectious hepatitis with gamma globulin. JAMA 129 (4):270–272 [Google Scholar]
  68. Heath PT (1998) Haemophilus influenzae type b conjugate vaccines: a review of efficacy data. Pediatr Infect Dis J 17 (9 Suppl):S117–122 [DOI] [PubMed] [Google Scholar]
  69. Heinz FX, Stiasny K, Holzmann H, Grgic-Vitek M, Kriz B, Essl A, Kundi M (2013) Vaccination and tick-borne encephalitis, central Europe. Emerg Infect Dis 19 (1):69–76. doi: 10.3201/eid1901.120458 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Hoffman SL, Vekemans J, Richie TL, Duffy PE (2015) The march toward malaria vaccines. Vaccine 33 Suppl 4:D13–23. doi: 10.1016/j.vaccine.2015.07.091 S0264-410X(15)01070-1 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Hsia DY, Lonsway M Jr., Gellis SS (1954) Gamma globulin in the prevention of infectious hepatitis; studies on the use of small doses in family outbreaks. N Engl J Med 250 (10):417–419. doi: 10.1056/NEJM195403112501004 [DOI] [PubMed] [Google Scholar]
  72. Innis BL, Snitbhan R, Kunasol P, Laorakpongse T, Poopatanakool W, Kozik CA, Suntayakorn S, Suknuntapong T, Safary A, Tang DB, et al. (1994) Protection against hepatitis A by an inactivated vaccine. JAMA 271 (17):1328–1334 [PubMed] [Google Scholar]
  73. Izurieta HS, Strebel PM, Blake PA (1997) Postlicensure effectiveness of varicella vaccine during an outbreak in a child care center. JAMA 278 (18):1495–1499 [PubMed] [Google Scholar]
  74. Jaberolansar N, Toth I, Young PR, Skwarczynski M (2016) Recent advances in the development of subunit-based RSV vaccines. Expert Rev Vaccines 15 (1):53–68. doi: 10.1586/14760584.2016.1105134 [DOI] [PubMed] [Google Scholar]
  75. Jenner E (1798) An inquiry into the causes and effects of the variolae vaccinae. Sampson Low, London [Google Scholar]
  76. Jenner E (1799) Further observations on the variolae vaccinae. Sampson Low, London [Google Scholar]
  77. Jenner E (1800) A continuation of facts and observations relative to the variolae vaccinae, or cowpox. Sampson Low, London [Google Scholar]
  78. Kapikian AZ, Mitchell RH, Chanock RM, Shvedoff RA, Stewart CE (1969) An epidemiologic study of altered clinical reactivity to respiratory syncytial (RS) virus infection in children previously vaccinated with an inactivated RS virus vaccine. Am J Epidemiol 89 (4):405–421 [DOI] [PubMed] [Google Scholar]
  79. Kaslow DC, Biernaux S (2015) RTS,S: Toward a first landmark on the Malaria Vaccine Technology Roadmap. Vaccine 33 (52):7425–7432. doi: 10.1016/j.vaccine.2015.09.061 S0264-410X(15)01337-7 [pii] [DOI] [PubMed] [Google Scholar]
  80. Kim DK, Bridges CB, Harriman KH, Advisory Committee on Immunization Practices AAIWG (2016) Advisory Committee on Immunization Practices Recommended Immunization Schedule for Adults Aged 19 Years or Older - United States, 2016. MMWR Morb Mortal Wkly Rep 65 (4):88–90. doi: 10.15585/mmwr.mm6504a5 [DOI] [PubMed] [Google Scholar]
  81. Kim HW, Canchola JG, Brandt CD, Pyles G, Chanock RM, Jensen K, Parrott RH (1969) Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am J Epidemiol 89 (4):422–434 [DOI] [PubMed] [Google Scholar]
  82. Kim JH, Excler JL, Michael NL (2015) Lessons from the RV144 Thai phase III HIV-1 vaccine trial and the search for correlates of protection. Annu Rev Med 66:423–437. doi: 10.1146/annurev-med-052912-123749 [DOI] [PubMed] [Google Scholar]
  83. Kirnbauer R, Booy F, Cheng N, Lowy DR, Schiller JT (1992) Papillomavirus L1 major capsid protein self-assembles into virus-like particles that are highly immunogenic. Proc Natl Acad Sci U S A 89 (24):12180–12184 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Klein NP, Bartlett J, Fireman B, Baxter R (2016) Waning Tdap Effectiveness in Adolescents. Pediatrics 137:1–9. doi: 10.1542/peds.2015-3326 [DOI] [PubMed] [Google Scholar]
  85. Klein NP, Bartlett J, Fireman B, Rowhani-Rahbar A, Baxter R (2013) Comparative effectiveness of acellular versus whole-cell pertussis vaccines in teenagers. Pediatrics 131:e1716–1722. doi: 10.1542/peds.2012-3836 [DOI] [PubMed] [Google Scholar]
  86. Koepke R, Eickhoff JC, Ayele RA, Petit AB, Schauer SL, Hopfensperger DJ, Conway JH, Davis JP (2014) Estimating the effectiveness of tetanus-diphtheria-acellular pertussis vaccine (Tdap) for preventing pertussis: evidence of rapidly waning immunity and difference in effectiveness by Tdap brand. J Infect Dis 210:942–953. doi: 10.1093/infdis/jiu322 [DOI] [PubMed] [Google Scholar]
  87. Kost RG, Straus SE (1996) Postherpetic neuralgia--pathogenesis, treatment, and prevention. N Engl J Med 335 (1):32–42. doi: 10.1056/NEJM199607043350107 [DOI] [PubMed] [Google Scholar]
  88. Kramarz P, France EK, Destefano F, Black SB, Shinefield H, Ward JI, Chang EJ, Chen RT, Shatin D, Hill J, Lieu T, Ogren JM (2001) Population-based study of rotavirus vaccination and intussusception. Pediatr Infect Dis J 20 (4):410–416 [DOI] [PubMed] [Google Scholar]
  89. Kreimer AR, Struyf F, Del Rosario-Raymundo MR, Hildesheim A, Skinner SR, Wacholder S, Garland SM, Herrero R, David MP, Wheeler CM , Costa Rica Vaccine Trial Study Group A, Gonzalez P, Jimenez S, Lowy DR, Pinto LA, Porras C, Rodriguez AC, Safaeian M, Schiffman M, Schiller JT, Schussler J, Sherman ME, Authors PSG, Bosch FX, Castellsague X, Chatterjee A, Chow SN, Descamps D, Diaz-Mitoma F, Dubin G, Germar MJ, Harper DM, Lewis DJ, Limson G, Naud P, Peters K, Poppe WA, Ramjattan B, Romanowski B, Salmeron J, Schwarz TF, Teixeira JC, Tjalma WA, Collaborators HPPIC-PI, Group GSKVCSS (2015) Efficacy of fewer than three doses of an HPV-16/18 AS04-adjuvanted vaccine: combined analysis of data from the Costa Rica Vaccine and PATRICIA Trials. Lancet Oncol 16 (7):775–786. doi: 10.1016/S1470-2045(15)00047-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Lal H, Cunningham AL, Godeaux O, Chlibek R, Diez-Domingo J, Hwang SJ, Levin MJ, McElhaney JE, Poder A, Puig-Barbera J, Vesikari T, Watanabe D, Weckx L, Zahaf T, Heineman TC (2015) Efficacy of an adjuvanted herpes zoster subunit vaccine in older adults. N Engl J Med 372 (22):2087–2096. doi: 10.1056/NEJMoa1501184 [DOI] [PubMed] [Google Scholar]
  91. Lee LA, Franzel L, Atwell J, Datta SD, Friberg IK, Goldie SJ, Reef SE, Schwalbe N, Simons E, Strebel PM, Sweet S, Suraratdecha C, Tam Y, Vynnycky E, Walker N, Walker DG, Hansen PM (2013) The estimated mortality impact of vaccinations forecast to be administered during 2011–2020 in 73 countries supported by the GAVI Alliance. Vaccine 31 Suppl 2:B61–72. doi: 10.1016/j.vaccine.2012.11.035 [DOI] [PubMed] [Google Scholar]
  92. Levin MJ, Smith JG, Kaufhold RM, Barber D, Hayward AR, Chan CY, Chan IS, Li DJ, Wang W, Keller PM, Shaw A, Silber JL, Schlienger K, Chalikonda I, Vessey SJ, Caulfield MJ (2003) Decline in varicella-zoster virus (VZV)-specific cell-mediated immunity with increasing age and boosting with a high-dose VZV vaccine. J Infect Dis 188 (9):1336–1344. doi:JID30798 [pii] 10.1086/379048 [DOI] [PubMed] [Google Scholar]
  93. Lieberman JM, Williams WR, Miller JM, Black S, Shinefield H, Henderson F, Marchant CD, Werzberger A, Halperin S, Hartzel J, Klopfer S, Schodel F, Kuter BJ (2006) The safety and immunogenicity of a quadrivalent measles, mumps, rubella and varicella vaccine in healthy children: a study of manufacturing consistency and persistence of antibody. Pediatr Infect Dis J 25 (7):615–622. doi: 10.1097/01.inf.0000220209.35074.0b 00006454-200607000-00010 [pii] [DOI] [PubMed] [Google Scholar]
  94. Liko J, Robison SG, Cieslak PR (2013) Priming with whole-cell versus acellular pertussis vaccine. N Engl J Med 368 (6):581–582. doi: 10.1056/NEJMc1212006 [DOI] [PubMed] [Google Scholar]
  95. Lopez AS, Guris D, Zimmerman L, Gladden L, Moore T, Haselow DT, Loparev VN, Schmid DS, Jumaan AO, Snow SL (2006) One dose of varicella vaccine does not prevent school outbreaks: is it time for a second dose? Pediatrics 117 (6):e1070–1077. doi:117/6/e1070 [pii] 10.1542/peds.2005-2085 [DOI] [PubMed] [Google Scholar]
  96. Medical Research Council (1956) Vaccination against whooping-cough; relation between protection in children and results of laboratory tests; a report to the Whooping-cough Immunization Committee of the Medical Research Council and to the medical officers of health for Cardiff, Leeds, Leyton, Manchester, Middlesex, Oxford, Poole, Tottenham, Walthamstow, and Wembley. Br Med J 2 (4990):454–462 [PMC free article] [PubMed] [Google Scholar]
  97. Miller AE (1980) Selective decline in cellular immune response to varicella-zoster in the elderly. Neurology 30 (6):582–587 [DOI] [PubMed] [Google Scholar]
  98. Misegades LK, Winter K, Harriman K, Talarico J, Messonnier NE, Clark TA, Martin SW (2012) Association of childhood pertussis with receipt of 5 doses of pertussis vaccine by time since last vaccine dose, California, 2010. JAMA 308:2126–2132. doi: 10.1001/jama.2012.14939 [DOI] [PubMed] [Google Scholar]
  99. Morrison VA, Johnson GR, Schmader KE, Levin MJ, Zhang JH, Looney DJ, Betts R, Gelb L, Guatelli JC, Harbecke R, Pachucki C, Keay S, Menzies B, Griffin MR, Kauffman CA, Marques A, Toney J, Boardman K, Su SC, Li X, Chan IS, Parrino J, Annunziato P, Oxman MN (2015) Long-term persistence of zoster vaccine efficacy. Clin Infect Dis 60 (6):900–909. doi: 10.1093/cid/ciu918 ciu918 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Morse JL (1913) Whooping-cough: A plea for more efficient public regulations relative to the control of this most serious and fatal disease. JAMA 60 (22):1677–1680 [Google Scholar]
  101. Murphy TV, Gargiullo PM, Massoudi MS, Nelson DB, Jumaan AO, Okoro CA, Zanardi LR, Setia S, Fair E, LeBaron CW, Wharton M, Livengood JR (2001) Intussusception among infants given an oral rotavirus vaccine. N Engl J Med 344 (8):564–572. doi: 10.1056/NEJM200102223440804 [DOI] [PubMed] [Google Scholar]
  102. Murray NE, Quam MB, Wilder-Smith A (2013) Epidemiology of dengue: past, present and future prospects. Clin Epidemiol 5:299–309. doi: 10.2147/CLEP.S34440 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Nair H, Nokes DJ, Gessner BD, Dherani M, Madhi SA, Singleton RJ, O’Brien KL, Roca A, Wright PF, Bruce N, Chandran A, Theodoratou E, Sutanto A, Sedyaningsih ER, Ngama M, Munywoki PK, Kartasasmita C, Simoes EA, Rudan I, Weber MW, Campbell H (2010) Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: a systematic review and meta-analysis. Lancet 375:1545–1555. doi: 10.1016/S0140-6736(10)60206-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Novartis Vaccines and Diagnostics (2015) BEXSERO® (Meningococcal Group B Vaccine) Package Insert Cambridge, MA [Google Scholar]
  105. Oxman MN (1995) Immunization to reduce the frequency and severity of herpes zoster and its complications. Neurology 45 (12 Suppl 8):S41–46 [DOI] [PubMed] [Google Scholar]
  106. Oxman MN, Levin MJ, Johnson GR, Schmader KE, Straus SE, Gelb LD, Arbeit RD, Simberkoff MS, Gershon AA, Davis LE, Weinberg A, Boardman KD, Williams HM, Zhang JH, Peduzzi PN, Beisel CE, Morrison VA, Guatelli JC, Brooks PA, Kauffman CA, Pachucki CT, Neuzil KM, Betts RF, Wright PF, Griffin MR, Brunell P, Soto NE, Marques AR, Keay SK, Goodman RP, Cotton DJ, Gnann JW Jr., Loutit J, Holodniy M, Keitel WA, Crawford GE, Yeh SS, Lobo Z, Toney JF, Greenberg RN, Keller PM, Harbecke R, Hayward AR, Irwin MR, Kyriakides TC, Chan CY, Chan IS, Wang WW, Annunziato PW, Silber JL (2005) A vaccine to prevent herpes zoster and postherpetic neuralgia in older adults. N Engl J Med 352 (22):2271–2284. doi:352/22/2271 [pii] 10.1056/NEJMoa051016 [DOI] [PubMed] [Google Scholar]
  107. Paavonen J, Jenkins D, Bosch FX, Naud P, Salmeron J, Wheeler CM, Chow SN, Apter DL, Kitchener HC, Castellsague X, de Carvalho NS, Skinner SR, Harper DM, Hedrick JA, Jaisamrarn U, Limson GA, Dionne M, Quint W, Spiessens B, Peeters P, Struyf F, Wieting SL, Lehtinen MO, Dubin G (2007) Efficacy of a prophylactic adjuvanted bivalent L1 virus-like-particle vaccine against infection with human papillomavirus types 16 and 18 in young women: an interim analysis of a phase III double-blind, randomised controlled trial. Lancet 369 (9580):2161–2170 [DOI] [PubMed] [Google Scholar]
  108. Pedraz C, Carbonell-Estrany X, Figueras-Aloy J, Quero J, Group IS (2003) Effect of palivizumab prophylaxis in decreasing respiratory syncytial virus hospitalizations in premature infants. Pediatr Infect Dis J 22 (9):823–827. doi: 10.1097/01.inf.0000086403.50417.7c [DOI] [PubMed] [Google Scholar]
  109. Peltola H, Kayhty H, Virtanen M, Makela PH (1984) Prevention of Hemophilus influenzae type b bacteremic infections with the capsular polysaccharide vaccine. N Engl J Med 310 (24):1561–1566. doi: 10.1056/NEJM198406143102404 [DOI] [PubMed] [Google Scholar]
  110. Plosker GL (2012) A/H5N1 prepandemic influenza vaccine (whole virion, vero cell-derived, inactivated) [Vepacel(R)]. Drugs 72 (11):1543–1557. doi: 10.2165/11209650-000000000-00000 [DOI] [PubMed] [Google Scholar]
  111. Plotkin SA (2014) The pertussis problem. Clin Infect Dis 58:830–833. doi: 10.1093/cid/cit934 [DOI] [PubMed] [Google Scholar]
  112. Polack FP (2007) Atypical measles and enhanced respiratory syncytial virus disease (ERD) made simple. Pediatr Res 62 (1):111–115. doi: 10.1203/PDR.0b013e3180686ce0 [DOI] [PubMed] [Google Scholar]
  113. Prevots DR, Sutter RW, Strebel PM, Weibel RE, Cochi SL (1994) Completeness of reporting for paralytic poliomyelitis, United States, 1980 through 1991. Implications for estimating the risk of vaccine-associated disease. Arch Pediatr Adolesc Med 148 (5):479–485 [DOI] [PubMed] [Google Scholar]
  114. Provost PJ, Hilleman MR (1979) Propagation of human hepatitis A virus in cell culture in vitro. Proc Soc Exp Biol Med 160 (2):213–221 [DOI] [PubMed] [Google Scholar]
  115. Provost PJ, Hughes JV, Miller WJ, Giesa PA, Banker FS, Emini EA (1986) An inactivated hepatitis A viral vaccine of cell culture origin. J Med Virol 19 (1):23–31 [DOI] [PubMed] [Google Scholar]
  116. Purcell RH, D’Hondt E, Bradbury R, Emerson SU, Govindarajan S, Binn L (1992) Inactivated hepatitis A vaccine: active and passive immunoprophylaxis in chimpanzees. Vaccine 10 Suppl 1:S148–151 [DOI] [PubMed] [Google Scholar]
  117. Rauh LW, Schmidt R (1965) Measles Immunization with Killed Virus Vaccine. Serum Antibody Titers and Experience with Exposure to Measles Epidemic. Am J Dis Child 109:232–237 [DOI] [PubMed] [Google Scholar]
  118. Rendi-Wagner P, Korinek M, Winkler B, Kundi M, Kollaritsch H, Wiedermann U (2007) Persistence of seroprotection 10 years after primary hepatitis A vaccination in an unselected study population. Vaccine 25:927–931. doi: 10.1016/j.vaccine.2006.08.044 [DOI] [PubMed] [Google Scholar]
  119. Reyes IS, Hsieh DT, Laux LC, Wilfong AA (2011) Alleged cases of vaccine encephalopathy rediagnosed years later as Dravet syndrome. Pediatrics 128:e699–702. doi: 10.1542/peds.2010-0887 [DOI] [PubMed] [Google Scholar]
  120. Richie TL, Billingsley PF, Sim BK, James ER, Chakravarty S, Epstein JE, Lyke KE, Mordmuller B, Alonso P, Duffy PE, Doumbo OK, Sauerwein RW, Tanner M, Abdulla S, Kremsner PG, Seder RA, Hoffman SL (2015) Progress with Plasmodium falciparum sporozoite (PfSPZ)-based malaria vaccines. Vaccine 33 (52):7452–7461. doi: 10.1016/j.vaccine.2015.09.096 S0264-410X(15)01386-9 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Roper M, Wassilak S, Tiwari T, Orenstein W (2013) Tetanus toxoid In: Plotkin SA, Orenstein WA, Offit PA (eds) Vaccines. 6th edn Saunders/Elsevier, Philadelphia, PA, pp 746–772 [Google Scholar]
  122. RTS-S Clinical Trials Partnership (2015) Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet 386 (9988):31–45. doi: 10.1016/S0140-6736(15)60721-8 S0140-6736(15)60721-8 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Sabchareon A, Wallace D, Sirivichayakul C, Limkittikul K, Chanthavanich P, Suvannadabba S, Jiwariyavej V, Dulyachai W, Pengsaa K, Wartel TA, Moureau A, Saville M, Bouckenooghe A, Viviani S, Tornieporth NG, Lang J (2012) Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: a randomised, controlled phase 2b trial. Lancet 380 (9853):1559–1567. doi:S0140-6736(12)61428-7 [pii] 10.1016/S0140-6736(12)61428-7 [DOI] [PubMed] [Google Scholar]
  124. Safaeian M, Porras C, Pan Y, Kreimer A, Schiller JT, Gonzalez P, Lowy DR, Wacholder S, Schiffman M, Rodriguez AC, Herrero R, Kemp T, Shelton G, Quint W, van Doorn LJ, Hildesheim A, Pinto LA (2013) Durable antibody responses following one dose of the bivalent human papillomavirus L1 virus-like particle vaccine in the Costa Rica Vaccine Trial. Cancer Prev Res (Phila) 6:1242–1250. doi: 10.1158/1940-6207.CAPR-13-0203 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Sako W (1947) Studies on pertussis immunization. J Pediatr 30 (1):29–40 [DOI] [PubMed] [Google Scholar]
  126. Pasteur Sanofi (2016) World’s First Public Dengue Immunization Program Starts in the Philippines. http://www.sanofipasteur.com/en/articles/World-s-First-Public-Dengue-Immunization-Program-Starts-in-the-Philippines.aspx. Accessed April 5 2016
  127. Santosham M, Reid R, Letson GW, Wolff MC, Siber G (1990) Passive immunization for infection with Haemophilus influenzae type b. Pediatrics 85 (4 Pt 2):662–666 [PubMed] [Google Scholar]
  128. Sato H, Sato Y (1990) Protective activities in mice of monoclonal antibodies against pertussis toxin. Infection and immunity 58 (10):3369–3374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Schiller JT, Hidesheim A (2000) Developing HPV virus-like particle vaccines to prevent cervical cancer: a progress report. J Clin Virol 19:67–74 [DOI] [PubMed] [Google Scholar]
  130. Schiller JT, Lowy DR (1996) Papillomavirus-like particles and HPV vaccine development. Semin Cancer Biol 7:373–382. doi: 10.1006/scbi.1996.0046 [DOI] [PubMed] [Google Scholar]
  131. Schmader KE, Oxman MN, Levin MJ, Johnson G, Zhang JH, Betts R, Morrison VA, Gelb L, Guatelli JC, Harbecke R, Pachucki C, Keay S, Menzies B, Griffin MR, Kauffman C, Marques A, Toney J, Keller PM, Li X, Chan IS, Annunziato P (2012) Persistence of the efficacy of zoster vaccine in the shingles prevention study and the short-term persistence substudy. Clin Infect Dis 55 (10):1320–1328. doi: 10.1093/cid/cis638 cis638 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Schneerson R, Rodrigues LP, Parke JC Jr., Robbins JB (1971) Immunity to disease caused by Hemophilus influenzae type b. II. Specificity and some biologic characteristics of “natural,” infection-acquired, and immunization-induced antibodies to the capsular polysaccharide of Hemophilus influenzae type b. J Immunol 107 (4):1081–1089 [PubMed] [Google Scholar]
  133. Schwarz TF, Leo O (2008) Immune response to human papillomavirus after prophylactic vaccination with AS04-adjuvanted HPV-16/18 vaccine: improving upon nature. Gynecol Oncol 110:S1–10. doi: 10.1016/j.ygyno.2008.05.036 [DOI] [PubMed] [Google Scholar]
  134. Seder RA, Chang LJ, Enama ME, Zephir KL, Sarwar UN, Gordon IJ, Holman LA, James ER, Billingsley PF, Gunasekera A, Richman A, Chakravarty S, Manoj A, Velmurugan S, Li M, Ruben AJ, Li T, Eappen AG, Stafford RE, Plummer SH, Hendel CS, Novik L, Costner PJ, Mendoza FH, Saunders JG, Nason MC, Richardson JH, Murphy J, Davidson SA, Richie TL, Sedegah M, Sutamihardja A, Fahle GA, Lyke KE, Laurens MB, Roederer M, Tewari K, Epstein JE, Sim BK, Ledgerwood JE, Graham BS, Hoffman SL (2013) Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 341 (6152):1359–1365. doi:science.1241800 [pii] 10.1126/science.1241800 [DOI] [PubMed] [Google Scholar]
  135. Sheridan SL, Ware RS, Grimwood K, Lambert SB (2012) Number and order of whole cell pertussis vaccines in infancy and disease protection. JAMA 308:454–456. doi: 10.1001/jama.2012.6364 [DOI] [PubMed] [Google Scholar]
  136. Shorvon S, Berg A (2008) Pertussis vaccination and epilepsy--an erratic history, new research and the mismatch between science and social policy. Epilepsia 49:219–225. doi: 10.1111/j.1528-1167.2007.01478.x [DOI] [PubMed] [Google Scholar]
  137. Simmons CP (2015) A Candidate Dengue Vaccine Walks a Tightrope. N Engl J Med 373 (13):1263–1264. doi: 10.1056/NEJMe1509442 [DOI] [PubMed] [Google Scholar]
  138. Slifka MK, Amanna I (2014) How advances in immunology provide insight into improving vaccine efficacy. Vaccine 32 (25):2948–2957. doi: 10.1016/j.vaccine.2014.03.078 S0264-410X(14)00459-9 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Stanaway JD, Shepard DS, Undurraga EA, Halasa YA, Coffeng LE, Brady OJ, Hay SI, Bedi N, Bensenor IM, Castaneda-Orjuela CA, Chuang TW, Gibney KB, Memish ZA, Rafay A, Ukwaja KN, Yonemoto N, Murray CJ (2016) The global burden of dengue: an analysis from the Global Burden of Disease Study 2013. Lancet Infect Dis. doi: 10.1016/S1473-3099(16)00026-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Stokes J Jr., Neefe JR (1945) The prevention and attenuation of infectious hepatitis by gamma globulin. JAMA 127 (3):144–145 [Google Scholar]
  141. Stokley S, Jeyarajah J, Yankey D, Cano M, Gee J, Roark J, Curtis RC, Markowitz L (2014) Human papillomavirus vaccination coverage among adolescents, 2007–2013, and postlicensure vaccine safety monitoring, 2006–2014--United States. MMWR Morb Mortal Wkly Rep 63:620–624 [PMC free article] [PubMed] [Google Scholar]
  142. Storsaeter J, Hallander HO, Gustafsson L, Olin P (1998) Levels of anti-pertussis antibodies related to protection after household exposure to Bordetella pertussis. Vaccine 16:1907–1916 [DOI] [PubMed] [Google Scholar]
  143. Strebel PM, Papania MJ, Fiebelkorn AP, Halsey NA (2013) Measles vaccines In: Plotkin SA, Orenstein WA, Offit PA (eds) Vaccines. 6th edn Saunders/Elsevier, Philadelphia, PA, pp 352–388 [Google Scholar]
  144. Tejedor JC, Merino JM, Moro M, Navarro ML, Espin J, Omenaca F, Garcia-Sicilia J, Moreno-Perez D, Ruiz-Contreras J, Centeno F, Barrio F, Cabanillas L, Muro M, Esporrin C, De Torres MJ, Caubet M, Boutriau D, Miller JM, Mesaros N (2012) Five-year antibody persistence and safety following a booster dose of combined Haemophilus influenzae type b-Neisseria meningitidis serogroup C-tetanus toxoid conjugate vaccine. Pediatr Infect Dis J 31 (10):1074–1077. doi: 10.1097/INF.0b013e318269433a [DOI] [PubMed] [Google Scholar]
  145. The FUTURE II Study Group (2007) Quadrivalent vaccine against human papillomavirus to prevent high-grade cervical lesions. N Engl J Med 356 (19):1915–1927 [DOI] [PubMed] [Google Scholar]
  146. The IMpact RSV Study Group (1998) Palivizumab, a Humanized Respiratory Syncytial Virus Monoclonal Antibody, Reduces Hospitalization From Respiratory Syncytial Virus Infection in High-risk Infants. Pediatrics 102 (3):531–537 [PubMed] [Google Scholar]
  147. Theeten H, Van Herck K, Van Der Meeren O, Crasta P, Van Damme P, Hens N (2015) Long-term antibody persistence after vaccination with a 2-dose Havrix (inactivated hepatitis A vaccine): 20 years of observed data, and long-term model-based predictions. Vaccine 33 (42):5723–5727. doi: 10.1016/j.vaccine.2015.07.008 [DOI] [PubMed] [Google Scholar]
  148. Tiwari T, Wharton M (2013) Diphtheria toxoid In: Plotkin SA, Orenstein WA, Offit PA (eds) Vaccines. 6th edn Saunders/Elsevier, Philadelphia, PA, pp 153–166 [Google Scholar]
  149. Tseng HF, Harpaz R, Luo Y, Hales CM, Sy LS, Tartof SY, Bialek S, Hechter RC, Jacobsen SJ (2016) Declining Effectiveness of Herpes Zoster Vaccine in Adults Aged >/=60 Years. J Infect Dis. doi:jiw047 [pii] 10.1093/infdis/jiw047 [DOI] [PubMed] [Google Scholar]
  150. Van Damme P, Thoelen S, Cramm M, De Groote K, Safary A, Meheus A (1994) Inactivated hepatitis A vaccine: reactogenicity, immunogenicity, and long-term antibody persistence. J Med Virol 44 (4):446–451 [DOI] [PubMed] [Google Scholar]
  151. Vaughn DW, Green S, Kalayanarooj S, Innis BL, Nimmannitya S, Suntayakorn S, Endy TP, Raengsakulrach B, Rothman AL, Ennis FA, Nisalak A (2000) Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity. J Infect Dis 181 (1):2–9. doi:JID990867 [pii] 10.1086/315215 [DOI] [PubMed] [Google Scholar]
  152. Vazquez M, LaRussa PS, Gershon AA, Steinberg SP, Freudigman K, Shapiro ED (2001) The effectiveness of the varicella vaccine in clinical practice. N Engl J Med 344 (13):955–960. doi:MJBA-441302 [pii] 10.1056/NEJM200103293441302 [DOI] [PubMed] [Google Scholar]
  153. Villar L, Dayan GH, Arredondo-Garcia JL, Rivera DM, Cunha R, Deseda C, Reynales H, Costa MS, Morales-Ramirez JO, Carrasquilla G, Rey LC, Dietze R, Luz K, Rivas E, Miranda Montoya MC, Cortes Supelano M, Zambrano B, Langevin E, Boaz M, Tornieporth N, Saville M, Noriega F, Group CYDS (2015) Efficacy of a tetravalent dengue vaccine in children in Latin America. N Engl J Med 372 (2):113–123. doi: 10.1056/NEJMoa1411037 [DOI] [PubMed] [Google Scholar]
  154. von Behring E, Kitasato S (1890) Ueber das zustandekommen der diphtherieimmunitat und der tetanus-immunitat bei thieren (On the realization of immunity in diphtheria and tetanus in animals). Deutsche Medizinische Wochenschrift 16:1113–1114 [Google Scholar]
  155. Walton LR, Orenstein WA, Pickering LK (2015) Lessons learned from making and implementing vaccine recommendations in the U.S. Vaccine 33 Suppl 4:D78–82. doi: 10.1016/j.vaccine.2015.09.036 [DOI] [PubMed] [Google Scholar]
  156. Ward JI, Broome CV, Harrison LH, Shinefield H, Black S (1988) Haemophilus influenzae type b vaccines: lessons for the future. Pediatrics 81 (6):886–893 [PubMed] [Google Scholar]
  157. Webster DP, Farrar J, Rowland-Jones S (2009) Progress towards a dengue vaccine. Lancet Infect Dis 9 (11):678–687. doi:S1473-3099(09)70254-3 [pii] 10.1016/S1473-3099(09)70254-3 [DOI] [PubMed] [Google Scholar]
  158. Werzberger A, Mensch B, Kuter B, Brown L, Lewis J, Sitrin R, Miller W, Shouval D, Wiens B, Calandra G, et al. (1992) A controlled trial of a formalin-inactivated hepatitis A vaccine in healthy children. N Engl J Med 327 (7):453–457. doi: 10.1056/NEJM199208133270702 [DOI] [PubMed] [Google Scholar]
  159. Wiedermann G, Kundi M, Ambrosch F, Safary A, D’Hondt E, Delem A (1997) Inactivated hepatitis A vaccine: long-term antibody persistence. Vaccine 15:612–615 [DOI] [PubMed] [Google Scholar]
  160. Williamson ED, Oyston PCF (2013) Plague vaccines In: Plotkin SA, Orenstein WA, Offit PA (eds) Vaccines. 5th edn Saunders/Elsevier, Philadelphia, PA, pp 493–503 [Google Scholar]
  161. Wilson A, Sharp M, Koropchak CM, Ting SF, Arvin AM (1992) Subclinical varicella-zoster virus viremia, herpes zoster, and T lymphocyte immunity to varicella-zoster viral antigens after bone marrow transplantation. J Infect Dis 165 (1):119–126 [DOI] [PubMed] [Google Scholar]
  162. Wolters KL, Dehmel H (1942) [Final investigations into tetanus prevention by active immunization]. Zeitschrift für hygiene und infektions krankheiten medizinische, mikrobiologie und viralogie 124:326–332 [Google Scholar]
  163. World Health Organization (2014) Global control and regional elimination of measles, 2000–2012. Wkly Epidemiol Rec 89 (6):45–52 [PubMed] [Google Scholar]
  164. Young MK, Nimmo GR, Cripps AW, Jones MA (2014) Post-exposure passive immunisation for preventing measles. Cochrane Database Syst Rev 4:CD010056. doi: 10.1002/14651858.CD010056.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Zhou F, Santoli J, Messonnier ML, Yusuf HR, Shefer A, Chu SY, Rodewald L, Harpaz R (2005) Economic evaluation of the 7-vaccine routine childhood immunization schedule in the United States, 2001. Arch Pediatr Adolesc Med 159 (12):1136–1144. doi:159/12/1136 [pii] 10.1001/archpedi.159.12.1136 [DOI] [PubMed] [Google Scholar]
  166. Zingher A (1924) Convalescent whole blood plasma and serum in prophylaxis of measles. JAMA 82 (15):1180–1187 [DOI] [PubMed] [Google Scholar]

RESOURCES