International Reviews of Immunology

ISSN: 0883-0185 (Print) 1563-5244 (Online) Journal homepage: http://www.tandfonline.com/loi/iiri20

Anti-infectious human vaccination in historical perspective Enrico D'Amelio, Simonetta Salemi & Raffaele D'Amelio To cite this article: Enrico D'Amelio, Simonetta Salemi & Raffaele D'Amelio (2015): Antiinfectious human vaccination in historical perspective, International Reviews of Immunology, DOI: 10.3109/08830185.2015.1082177 To link to this article: http://dx.doi.org/10.3109/08830185.2015.1082177

Published online: 25 Nov 2015.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=iiri20 Download by: [University of California Santa Barbara]

Date: 26 November 2015, At: 23:37

INTERNATIONAL REVIEWS OF IMMUNOLOGY http://dx.doi.org/./..

REVIEW

Anti-infectious human vaccination in historical perspective Enrico D’Amelioa,∗ , Simonetta Salemic,∗ , and Raffaele D’Ameliob,c

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

a Via Felice Grossi Gondi Rome, Italy; b Sapienza University of Rome, Department of Clinical and Molecular Medicine, Via di Grottarossa Rome, Italy; c S. Andrea University Hospital, Via di Grottarossa Rome, Italy

ABSTRACT

ARTICLE HISTORY

A brief history of vaccination is presented since the Jenner’s observation, through the first golden age of vaccinology (from Pasteur’s era to 1938), the second golden age (from 1940 to 1970), until the current period. In the first golden age, live, such as Bacille Calmette Guérin (BCG), and yellow fever, inactivated, such as typhoid, cholera, plague, and influenza, and subunit vaccines, such as tetanus and diphtheria toxoids, have been developed. In the second golden age, the cell culture technology enabled polio, measles, mumps, and rubella vaccines be developed. In the era of modern vaccines, in addition to the conjugate polysaccharide, hepatitis A, oral typhoid, and varicella vaccines, the advent of molecular biology enabled to develop hepatitis B, acellular pertussis, papillomavirus, and rotavirus recombinant vaccines. Great successes have been achieved in the fight against infectious diseases, including the smallpox global eradication, the nearly disappearance of polio, the control of tetanus, diphtheria, measles, rubella, yellow fever, and rabies. However, much work should still be done for improving old vaccines, such as BCG, anthrax, smallpox, plague, or for developing effective vaccines against old or emerging infectious threats, such as human-immunodeficiency-virus, malaria, hepatitis C, dengue, respiratory-syncytial-virus, cytomegalovirus, multiresistant bacteria, Clostridium difficile, Ebola virus. In addition to search for innovative and effective vaccines and global infant coverage, even risk categories should adequately be protected. Despite patients under immunosuppressive therapy are globally increasing, their vaccine coverage is lower than recommended, even in developed and affluent countries.

Accepted  July  KEYWORDS

History; infections; vaccinations; vaccines; vaccinology

Introduction Among the possible means to control infectious diseases, clean water and vaccination are, in the order, the most effective [1]. The results obtained through vaccination are easily demonstrated by both, epidemiological data [2,3] and the consideration that, for the first time in the history of mankind, one infectious disease, such as smallpox, has been eradicated following a clear strategy of vaccination campaign [4]. Thus, a brief history of vaccination allows this practice be adequately valued in the control of infectious diseases, as accurate expression of the scientific advances in the fields of microbiology, immunology and molecular biology. Moreover,

CONTACT Raffaele D’Amelio raff[email protected] Sapienza University of Rome, Department of Clinical and Molecular Medicine, S. Andrea University Hospital, Via di Grottarossa ,  Rome, Italy ∗ These Authors contributed equally ©  Taylor & Francis Group, LLC

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

2

E. D’AMELIO ET AL.

the antigen changes induced by the continuous mutual interrelationship between microorganisms and immune system, which are often unknown, are witnessed by the difficulty in developing effective vaccines towards parasites [5], mycobacteria [6], and RNA viruses [7]. The brief history of vaccination will be developed since the origin (the discovery of the Variola vaccine by Jenner and the introduction of the word “vaccination” [8]), through the first golden age (the period from Pasteur’s era to 1938), the second golden age (1940–1970, characterized by the discovery of cell culture technology), until the rationally designed, modern vaccines [9]. During the first golden age of vaccinology live attenuated [rabies, tuberculosis and yellow fever], inactivated [typhoid, cholera, plague and pertussis] and subunit [tetanus and diphtheria] vaccines have been developed. During the second golden age, instead, cellculture technology has allowed polio, measles, mumps, and rubella vaccines be developed. In spite of the undeniable merits of vaccines in the fight against infectious diseases, for a long time they have been developed empirically. Only recently, in consideration of the difficulties in obtaining a protective vaccine against human immunodeficiency virus (HIV), malaria or tuberculosis, the need to identify the correlates of protection for the different microorganisms has been underscored [10]. By this knowledge, vaccines and adjuvants able to selectively stimulate the protective immune response may be developed.

The origin Since the 10th century AD, trials in China to prevent/mitigate smallpox were reported. This could be obtained by the so called “variolation,” that is the inoculation of scab material from patients suffering of smallpox to healthy people, with the hope of inducing a protective mild disease. This practice, however, was not free from the risk of provoking smallpox [3]. At the end of the 18th century, the English physician Edward Anthony Jenner tried a new strategy, by inoculating humans with the material from cowpox blister instead of the material from human scabs. The trial was successful, by achieving an effective protection with a reasonable low risk of developing disease. This strategy was based on the observation that the farmers milking the cows infected by cowpox only caught a localized mild disease, but resulted protected towards the human smallpox. These data were published in 1798 and opened the era of vaccination [11]. The general practice of inoculating attenuated/inactivated microorganism to prevent infectious diseases was, in fact, named “vaccination” after the Latin word “vacca” which means “cow” [12]. It should be underlined that the association realized by Jenner of a sort of cross-protection between cowpox and smallpox is even more brilliant considering that it happened in a period when: (1) the germ theory was not yet established; (2) the reasons for the disease-spreading phenomenon were quite unclear; (3) the awareness of the immune system existence, with its operating rules, such as cross-protection, was lacking. Today we know that smallpox is caused by the Variola virus (major or minor according to the severity of the induced disease), pertaining to the Orthopoxvirus genus of the Poxviridae family, which also includes Vaccinia, Cowpox, and Monkeypox. All of them are potentially pathogens for human species. Vaccinia virus is often confused with Cowpox virus, based on the Jenner’s report that cowpox had been used, but they represent two distinct species of the Orthopoxvirus genus. During the 20th century, it was established that all available smallpox vaccines were based on Vaccinia virus [3]. Despite the quick success of vaccination in different countries, including the Americas and Asia, in addition to Europe, Jenner was even contested, also in relation to the priority of his discovery [13]. The smallpox in the 18th century killed an estimated 400,000 Europeans each year, was responsible of over 30% of all blindness and of the death of 20–60% of all infected patients. Still

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

3

in 1967 it was estimated to be responsible of 15 million cases and 2 million deaths [14,15]. This vaccine, although quite reactogenic, has been very effective and systematically administered in many world countries. Already in 1812, the US War Department ordered that vaccination be substituted for variolation to prevent smallpox [16]. However, the limit for the worldwide vaccination was represented by the reduced viability of the liquid vaccine lymph during the transports, mainly to tropical areas. A real advancement, in the course of the first decades of the twentieth century, was achieved, therefore, with the dehydration before and lyophilization afterwards (during the First World War) of the lymph. Nevertheless, despite in the 50s of the last century smallpox was eradicated in many areas of Europe and North America, still in 1958 its consequences were catastrophic in 63 countries, thus stimulating the World Health Assembly (WHA) to set in motion the process of worldwide eradication [4]. The aggressive eradication campaign, planned by the World Health Organization (WHO) and headed by Donald Ainslie Henderson, started in 1967 and, after a decade, in 1977 the last case of natural smallpox was discovered in Somalia. The approved budget for eradication was $ 2.4 million/year for 10 years, compared with over $ 2.5 billion for the failed malaria eradication campaign between 1957 and 1975 [4]. After the successful eradication campaign, the WHO announced the disease eradication and recommended to cease vaccination worldwide. However, following the dramatic episode in the United States in 2001 of the anthrax-contaminated letters, lack of immunization for smallpox has been considered a vulnerability towards a bioterrorism threat. A renewed interest for a safer and at least equally effective vaccine has been, therefore, observed after over two decades since smallpox eradication. Smallpox has, in fact, been included among the most dangerous, category A, possible biological weapons [17]. This new situation induced the US Department of Defense (DoD) to reinstitute large-scale vaccination for the military personnel. Over 1,500,000 in the DoD and 39,000 individuals in the Department of Health and Human Services were, therefore, vaccinated in 2002 [18]. The possible future vaccines for smallpox include attenuated strains, such as the Modified Vaccinia virus Ankara (MVA), which has been attenuated over serial (> 500) passages on chicken embryo fibroblasts, or the fourth generation vaccines, represented by subunit, recombinant antigens, expressed by proteins or DNA genes [3].

The first golden age of vaccinology Despite the success in the fight against smallpox, the observation by Edward Jenner remained isolated for nearly a century. Times too early and lack of association between birth of vaccination and knowledge of microbiology and immunology prevented the extension to other infectious diseases of the principle that inoculation of an attenuated microorganism could protect from natural infection. This happened in the second half of the nineteenth century, when the chemist Louis Pasteur approached biology with scientific method, thus opening the era of microbiology and immunology, together with the contemporaneous Robert Koch. The first golden age spans a period of time ranging from Pasteur’s era to 1938.

Pasteur’s era In 1878, Pasteur discovered the bacterial etiology of the so-called fowl cholera—the agent was later named Pasteurella multocida in his honor. In the following year, the casual observation that the old bacterial cultures resulted attenuated [19] paved the way for applying vaccination to other infectious diseases [20,21]. The next interest of Pasteur was addressed to anthrax, the

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

4

E. D’AMELIO ET AL.

bacterial origin of which had been discovered by Robert Koch, who failed to obtain an attenuated bacterial strain in spite of serial subcultures [19]. Pasteur succeeded by preventing spore formation, thus a vaccine was prepared and tested in 1881 on animals; all vaccinated animals survived in good health following inoculation of virulent bacteria, whereas all the nonvaccinated animals died or fell severely ill in 2–3 days [22]. During the last century, anthrax human vaccines have been developed, either live in the former Soviet Union or acellular, including the protective antigen of the anthrax toxin, in the UK and United States. Despite the demonstrated safety and efficacy, the need for an innovative anthrax vaccine is highly felt, even in consideration that Bacillus anthracis, the etiological agent of anthrax, has been included among the most dreadful category A biological agents [17]. Thus, vaccines based on recombinant protective antigen of anthrax toxin are in phase 2 clinical trials [23]. Afterwards Louis Pasteur turned to the study of rabies, an almost invariably lethal disease for humans even today. He succeeded in attenuating the virus through different methods, including either the passage from dogs to monkeys or the desiccation of rabbit spinal cord followed by air exposure for 15 days [24]. The vaccine obtained by the second method was successfully tested for the first time in July 1885 on a 9-year-old boy bitten by a rabid dog [12]. Profiting from the rabies long incubation period, vaccination resulted protective in an already infected patient. It enabled to consider the vaccination use not only for the infection prevention in healthy people, but also for the treatment of already infected patients, provided that the incubation period be long enough. In addition to rabies, in which therapeutic vaccination in case of suspected bites is still used, a post-exposure protection has also been observed for hepatitis B (HB), hepatitis A (HA), measles, and varicella vaccines [25] and this approach has been tried, even though unsuccessfully as yet, in the search of a therapeutic vaccine for HIV. Soon after Pasteur’s discovery of the possibility to attenuate the bacterial virulence, other Authors took advantage of methods able to physically or chemically completely inactivate bacteria as a new vaccine source [26]. Thus, the first typhoid [27,28], cholera [29], and plague [30] vaccines, all consisting of killed whole bacterial bodies, were developed by the end of 19th century [31]. In contrast with the live attenuated, the inactivated vaccines are frequently safer, but generally less effective. Typhoid and cholera vaccines have, in fact, been replaced by more effective new vaccines, whereas the need for a more effective innovative vaccine for plague is felt principally in the light of the bioterrorism threat, considering that even Yersinia pestis, the etiological agent of plague, has been included among the most dreadful category A agents [17]. The new typhoid and cholera vaccines will be later reported, whereas plague has been updated by adding the live and the subunit (F1 and V) vaccines. However, studies analyzing the efficacy of the single plague vaccines or comparing the different vaccines are lacking, thus information is only coming from animal models [32]. A recombinant subunit plague vaccine is in advanced development, even though efficacy will only be explored in animal models before licensing, for ethical and practical reasons [33]. The great scientific credit earned and the important school founded by Pasteur enabled the network of Pasteur Institutes in France and in overseas African and Asian French colonies be organized. It has provided a substantial contribution to the development of vaccinology, as recently named the science which deals with the study of vaccines [34]. Tuberculosis In the first decades of the 20th century two new live as well as the bacterial-derived toxoid vaccines were developed. The live bacterial anti-tubercular vaccine Bacille Calmette-Guérin (BCG), after Albert Calmette and Camille Guérin, was for the first time successfully used in

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

5

1921, after over 25 years of studies, interrupted by the first World War. BCG was orally administered to a 3-day baby, born to a mother who had died of pulmonary tuberculosis one day after delivery [35]. Mycobacterium tuberculosis, the etiologic agent of human tuberculosis, had been discovered in 1882 by Robert Koch [36], whose efforts to develop a vaccine had been, however, unsuccessful. Tuberculosis was very feared because widespread and burdened with high mortality, of the order of 300/100,000 in 1895 in Lille (France). In the same year Calmette, as Director of the Pasteur Institute in Lille, started his studies on tuberculosis, which eventually led to vaccine development [35]. Calmette and Guérin used, for vaccine preparation, Mycobacterium bovis instead of Mycobacterium tuberculosis, attenuated by several passages in a culture medium supplemented with sterile beef bile. They took advantage, in fact, of both the Jenner’s experience on the use of a different species related pathogen and Pasteur’s experience on the attenuation of microorganisms. Since 1921, BCG has been safely administered to approximately 4 billion subjects [35,37,38] and included among the vaccines provided by the Expanded Program on Immunization (EPI) of the WHO. In spite of the Lubeck disaster in 1930 (the administration of a BCG contaminated with virulent M. tuberculosis to 250 children caused 73 deaths and 135 infections who recovered [38]), and the incomplete protection provided by the vaccine, quoted in the range 0–80%, depending on the type of disease and population [39], BCG is still largely used. In particular, BCG is very effective against miliary and meningeal tuberculosis in < 5-year-old children, especially if not previously infected with M. tuberculosis or sensitized with environmental mycobacteria [40]. Although neonatal vaccination is cost-effective in high-burden countries [41], adult vaccination only provides little protection, especially against pulmonary tuberculosis, with a possible negative influence of decreasing latitude [40]. Moreover, it cannot be used as a booster, when the immunity wanes, generally after a decade [42], does not eradicate latent tuberculosis and does not prevent subsequent tuberculosis [43]. In HIV-infected infants evidence of protection exerted by BCG is limited and the risk of disseminated BCG very high (1100–4170 per million in HIV-infected infants versus 70% [43]. Consequently, WHO stated that HIV represents a full contraindication for BCG vaccination in infants, even though at high risk for tuberculosis infection [44]. Conversely, BCG has demonstrated to be protective towards other mycobacterial diseases, such as leprosy and Buruli ulcer, and capable to improve atopic disorders, nematode infections, bladder cancer, melanoma, and immune response to other vaccines [45,46]. Although BCG is only a partial solution to the problem of tuberculosis prevention, after nearly a century since its discovery no more effective vaccines have still been developed. It underlines the complex biology of the microorganism and of its interaction with the host. New live recombinant vaccines substituting BCG, or adjuvanted recombinant proteins, or viral vector expressing antigens, able to boost the immune response induced by BCG priming [prime-boost strategy], are currently under development [9]. Lack of availability of an effective vaccine, however, has influenced the poor disease control. This has been underlined by the WHO in the last decade of last century [47], by defining tuberculosis a global emergency, with about one third of the world’s population infected, a current annual estimate of nearly 9 million new cases and 1.7 million deaths [48]. Yellow fever The yellow fever is a viral disease transmitted by infected Aedes aegypti and Haemagogus species mosquitoes, endemic in Sub-Saharan Africa and tropical South America. It is responsible for 200,000 clinical cases and 30,000 deaths annually, according to WHO estimates. The live viral vaccine against yellow fever was prepared by Theiler and Smith, by attenuating the

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

6

E. D’AMELIO ET AL.

virus through passages in mice and chick embryos [49]. This vaccine, derived from an African patient (Asibi was his name, thus it was denominated the Asibi strain) with a relatively mild form of the disease, was successfully inoculated in a Macacus rhesus to reproduce the disease. Thus, the first laboratory model of the disease was obtained. Afterwards, this strain was attenuated through serial passages in the mouse brain, according to the Pasteurs method of attenuating rabies virus through passages in nonhost nervous tissue [50]. Meanwhile, Andrew Watson Sellards, from the department of tropical medicine at Harvard, was in Dakar, Senegal, to study a local outbreak of yellow fever and he heard of the success with Asibi strain. He joined Constant Mathis, the Director of the local Pasteur Institute, and Jean Laigret, who was responsible of the health defense to control the outbreak. Laigret had injected the blood from a patient with mild yellow fever into a rhesus monkey, thereby producing a severe form of the disease. This strain, attenuated through serial passages on mice brain and described by Sellards, Mathis and Laigret [51], was named French strain as opposed to Asibi strain. Both have been used as vaccines. The Asibi strain was used together with serum antibodies to reduce its virulence, whereas the French strain, which could induce fever and central nervous system reactions, was used in combination with smallpox vaccine. Both, in fact, could be administered by scarification. The two vaccine strains coexisted until 1982, when the French strain ceased to be produced. During the passages for attenuation, in fact, both strains, while reducing hepatotropism, increased neurotropism. However, only the Asibi strain, after the hundredth passage in nervous tissue-deprived chick embryo, showed a markedly reduced neurovirulence, being unable to kill intracerebrally-inoculated mice. Denominated 17D strain [52], it was used by Fred L. Soper, of the Rockefeller Foundation, in 1938 in a large vaccination campaign in Brazil, on nearly one million inhabitants, thereby proving its safety and efficacy [53]. It is still used, mainly when traveling to endemic countries, for which vaccination is compulsory, according to the international health regulations [54]. Despite the search for a safe and effective yellow fever vaccine has been a joint effort of different researchers, only Theiler was awarded the Nobel Prize in Physiology and Medicine in 1951, for his discoveries on yellow fever and how to combat it. Recently, after many decades of successful use of this vaccine, some insights have been provided on how it works. It strongly stimulates, in fact, innate immunity, through the activation of different Toll-like receptors on dendritic cells. These cells release pro-inflammatory cytokines, as interleukin (IL)12p40, IL6 and interferon (IFN)α, eventually leading to a mixed T helper (Th)1/Th2 cytokine profile and antigen-specific CD8+ T cells [55]. More recently, the same group [56] and another [57], independently and successfully, applied the systems biology approach to this vaccine for the first time. Briefly, they: (1) confirmed the anti-viral response, with activation of IFN type I pathway; (2) observed the activation of genes involved in the regulation of complement and inflammasome; (3) identified the genes able to predict neutralizing antibodies and specific CD8+ T-cell response. Tetanus, diphtheria, botulinum, and toxoid vaccines The recognition in 1888 by Émile Roux and Alexandre Yersin [58] and in 1890 by Faber [59] of the relevance for virulence of the toxins, in Corynebacterium diphtheriae and Clostridium tetani [60], led to vaccine preparation. Vaccines have been performed through the toxoids, that is the formaldehyde-inactivated toxins, by Gaston Ramon in 1923 and 1926, respectively [61,62]. Toxoids maintain an acceptable antigenicity, while losing the toxin virulence. These vaccines resulted very effective, thus inducing an excellent control of diphtheria and tetanus. At the end of the second World War, a formaldehyde-induced toxoid vaccine for protection towards Clostridium botulinum toxins was even developed in the United States. The toxins are

INTERNATIONAL REVIEWS OF IMMUNOLOGY

7

seven, from A to G, four of which (A, B, E and F) cause naturally occurring human botulism, with the other ones causing human botulism only at high doses. Consequently, the vaccine was before developed as bivalent (A/B), afterwards as pentavalent (A-E). It was only released as investigational new drug (IND) for at-risk-workers and the military. Clostridium botulinum has, in fact, been included among the most virulent category A biological agents [17]. More recently also recombinant adjuvanted molecules, corresponding to a part of toxin or to the whole molecule devoid of its catalytic activity, have been developed and tested as possible vaccines [63].

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

Pertussis In this period also whole-cell pertussis vaccine has been prepared, through Bordetella pertussis inactivation [64], usually by heating. The vaccine, mostly combined with tetanus and diphtheria toxoids (the trivalent DTP vaccine has been included in the EPI by the WHO), proved effective, but also able to induce local and systemic adverse events. The former have been especially observed in adolescents and adults, for whom it is, therefore, recommended at a reduced dose [65]. The introduction of the whole-cell inactivated vaccine in the forties of the last century in the United States has enabled annual cases, which were nearly 200,000 in the pre-vaccine era, be gradually reduced until approximately 1,000 in 1976. In the developing countries, pertussis is still a problem, with 89,000 estimated deaths in 2008 [66]. However, in the United States, starting from the eighties of the last century, a progressive increase of annual cases, even in adolescents and adults, for still unclear reasons, has been observed, with over 48,000 cases in 2012 [67]. Among the developed countries, increased case incidence seems to be observed in the United States only. In Italy, an epidemiological study performed in the 90s of the last century in the military enabled to exclude a situation like the one observed in the United States [68]. The need for a systematic administration of an additional booster vaccine dose in the USA adulthood should therefore be evaluated and in case recommended. Influenza In the first half of the 20th century, the viral nature of influenza was discovered, the influenza viruses characterized and cultivated in chick embryo tissue [69]. Thus, the first live influenza vaccines were developed and tested in the 30s of the last century [70,71]. A few years later the first killed influenza vaccine, effective although reactogenic, because poorly purified, was developed [72]. Since then, the inactivated influenza vaccine has been purified, in order to only include subunits, represented by the viral proteins necessary for host infection (haemagglutinin [HA]) and viral spread (neuraminidase [NA]). These may slightly (drift) or profoundly (shift) mutate, each year or with unpredictable periodicity, respectively. Thus, multivalent vaccines, including the subunits from circulating strains, have been developed. Recently, a live nasal influenza vaccine has been licensed in the United States and comparatively studied with the inactivated one through the systems vaccinology approach. The live vaccine, like the yellow fever vaccine, was able to stimulate the type I IFN response, but it was less effective in stimulating a systemic antibody response. Interestingly, the relevance of the gene product calcium/calmodulin-dependent kinase IV in the negative regulation of antiinfluenza antibody response, never appreciated before, has been observed [73]. The two vaccines have similar effectiveness in preventing influenza-like illness and influenza/pneumonia events in healthy adults [74]. However, they work through different mechanisms: live vaccine,

8

E. D’AMELIO ET AL.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

in fact, in analogy with natural infection and live polio vaccine, stimulates immune system at mucosal level, with consequent local and systemic immune response [75], including specific cell-mediated immunity [76]. Inactivated vaccine, instead, especially works through the stimulation of circulating antibody-secreting cells [73]. Whatever the route of administration and the mechanism of action, influenza vaccines, deriving from an RNA virus, are characterized by high antigenic variability, which reduces the effectiveness and makes the annual vaccine repetition a need. However, experimental and clinical trials have recently been performed for “universal” vaccines. They may be obtained by including in the vaccine composition viral invariant proteins, such as the M2 ion channel protein and the stalk domain (HA2) of the haemagglutinin [75,77].

Cell culture technology and the second golden age of vaccinology In the middle of the 20th century, the cell culture method was adapted to grow viruses. Moreover, it was also recognized to be able to fortuitously select less virulent strains, thus fit to directly develop viral vaccines [31,78]. For their discovery of the ability of poliomyelitis viruses to grow in culture of various types of tissue John F. Enders, Frederick S. Weller and Thomas H. Robbins were awarded the Nobel Prize for Physiology and Medicine in 1954. This achievement enabled to isolate, maintain and attenuate a viral vaccine strain in the laboratory, instead of to be forced to look for an animal model, as performed before. In the second half of the 20th century different live attenuated or inactivated viral vaccines have, therefore, been developed, including polio, measles, rubella, and mumps [31]. In this period, mainly during World War II, the need to protect the military represented an important stimulus to produce large quantities of the inactivated typhus and influenza vaccines and to develop the inactivated Japanese B encephalitis vaccine [79]. The mosquitotransmitted etiologic agent of Japanese encephalitis is a flavivirus, similar to West Nile virus. More recent Japanese encephalitis virus vaccines are cell cultured, live attenuated, and highly safe and immunogenic. The annual incidence is of 50,000–175,000 cases, with a mortality of 20–30% and permanent sequelae in 30–50% of the survivors. Even a recombinant, live attenuated, vectored chimeric yellow fever-Japanese encephalitis vaccine has been developed and is commercially available in Australia and Thailand [80]. Poliomyelitis The history of the development of the polio vaccines, which are included among the most significant advances of the last century, is worth being briefly reported for their relevance in the control of this dreaded disease. Until the middle of the last century in many world countries periodical poliomyelitis outbreaks struck children, adolescents and adults, with a mortality rate of paralytic cases which was 2–5% for children and 15–30% for adults [81]. During the first half of the century, it was established that polio is caused by viruses, which may infect the host by oral route [82] and that the viral strains are three [83]. Moreover, it was also observed that oral infection was accompanied by a transient intestinal carrier state and a viremic phase. The induced specific antibodies were found to be protective. This information was crucial for designing inactivated and oral vaccines, which were thus developed [84,85,86]. First Jonas Salk developed a formaldehyde-inactivated parenteral vaccine [87], which was adopted by United States in 1955 and largely used [88]. This vaccine led to a decrease of the incidence of paralytic cases from 13.9/100,000 in 1954 to 0.8/100,000 in 1961 [89], although the accident of Cutter Laboratories, Berkeley, had caused 260 cases of poliomyelitis and 10 deaths, due

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

9

to a failure of vaccine virus inactivation. In 1987, an inactivated polio vaccine of enhanced potency was licensed in the United States [90]. The history of oral polio vaccine, instead, consists of the independent efforts of at least three groups of researchers in the United States, led by Hilary Koprowsky at the Wistar Institute in Philadelphia, Herald Cox at the Lederle Laboratories and Albert Sabin, at the Children’s Hospital Research Foundation in Cincinnati. First Koprowsky developed a type 2 strain rodent-adapted oral vaccine [91]. A few years later, Herald Cox developed another oral polio vaccine, by using viral strains, some of which related to the strain of Koprowsky, who had originally been at Lederle Laboratories, together with Cox. Albert Sabin started his work on oral polio vaccine in 1953 and chose polio strains cultivated in cynomolgus monkey kidney, driven by the prior work of Ender’s group [92]. He obtained a live attenuated oral vaccine, which was considered less neurotropic than the other two oral vaccines, thus licensed at the beginning of the 60s of the last century [93]. The easy oral administration, which is the same route of the natural infection (polioviruses are, in fact, enteroviruses), the consequent consideration of a double level of defense, mucosal and circulatory, and the long-lasting protective immune response [92], in addition to its low cost, determined the success of the Sabin’s vaccine. It was, in fact, adopted in many world countries and inserted in the EPI by the WHO. In 1972 Sabin donated his vaccine strains to the WHO [92]. Although the live attenuated vaccine may very rarely induce paralytic polio in vaccinated subjects, generally for a reverse to virulence of attenuated viruses, particularly the type 3 (patients affected by humoral immunodeficiency are at special risk), the vaccine role in polio control has been so relevant that the current WHO plan foresees a polio eradication by 2018 [94]. Should this goal be achieved, polio would be the second eradicated infective disease, after smallpox, both as a consequence of vaccine use.

Adenoviruses In the same period, at the Walter Reed Army Institute of Research in United States, Maurice Hilleman, a scientist with a central and unprecedented role in vaccinology for the high number of developed vaccines, discovered the adenoviruses and produced a killed vaccine, which was licensed in 1958 for pediatric use [79]. A live oral bivalent adenovirus vaccine was approved for the use in the military only and administered since 1971 until 1999, when it was interrupted for more than 10 years and restored in 2011 [95].

Measles, mumps and rubella In the 50s of the last century, Enders and colleagues succeeded in preparing the attenuated Edmonston B viral strain for measles vaccine, which was licensed in the United States in 1963 [96]. Maurice Hilleman and colleagues, instead, could isolate and develop the attenuated mumps vaccine strain Jeryl Lynn, after Hilleman’s daughter [97], as well as a live attenuated rubella vaccine strain [98]. The efficacy of the trivalent live attenuated measles/mumps/rubella (MMR) vaccine in male adults was calculated to be nearly 95% for measles and rubella [99], lower for mumps, probably as a consequence of poor viral attenuation [100]. Measles vaccine has been included in the EPI by the WHO. However, measles cases are still too many and on the increase from 2010 to 2011 in Europe, Africa, South East Asia, and Eastern Mediterranean Region [101], thus inducing the WHO to revise and postpone the WHA 2010 measles eradication goal to be achieved by 2015. The Global Measles and Rubella Strategic Plan, in fact, considers measles elimination in at least five of six WHO regions by 2020. MMR coverage

10

E. D’AMELIO ET AL.

has been, at least temporarily, reduced in many countries, as a consequence of a presumed association MMR/autism [102,103], later on retracted by Lancet [104].

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

The modern era of vaccines—the advent of molecular biology This period corresponds to the last three decades of the last century and is principally marked by the advent of molecular biology. However, not all the vaccines produced in those years have been obtained by molecular biology. Some vaccines have, in fact, been performed according to traditional methods. Sometimes both, recombinant and traditional vaccines have been developed towards the same microorganism (this is the case for HB virus, rotavirus and cholera) and may coexist (rotavirus and cholera only). Moreover, an increasing attention to the study of vaccine immune response has also been provided, as demonstrated by the conjugate polysaccharide vaccines. HB vaccines and recombinant vaccines The HB virus was first identified by Baruch S. Blumberg in 1964 in an Australian Aborigenal person and called Australia antigen, later identified as the viral envelope protein [105]. In 1968, Alfred Prince associated this antigen with the serum hepatitis[106]. Baruch Blumberg in 1976 was awarded, together with D. Carleton Gajdusek, the Nobel Prize in Physiology and Medicine for their discoveries concerning the replication mechanism and the genetic structure of the viruses. HB virus is transmitted from mother to son, through infected blood or blood products and through sexual intercourse. More than 90% of infected people recover spontaneously, whereas approximately 10% proceed to chronic state. This especially happens in infants, who cannot clear the virus for the immune system immaturity. HB virus is responsible for an estimated 240 million subjects chronically infected in the world, about 780,000 of whom are estimated to yearly die as a consequence of acute infection, cirrhosis or hepatocellular carcinoma [107]. Thus vaccine prevention is pivotal. The first vaccine was obtained by Maurice Hilleman in Merck from seropositive plasma, chemically treated to inactivate any possible virulence [110]. However, the contemporaneous advances in molecular biology led to the development of the first recombinant vaccine represented by the engineered HB surface antigen, the gene of which had been inserted in a yeast, which was licensed in 1986 [108,109]. This innovative vaccine replaced the plasma-derived [110], which had been licensed in 1981 and proven to be safe and effective [79]. Currently, at least 183/194 WHO Member States vaccinate their infants for HB, thus it is predictable that HB virus infection will be largely controlled within the next few decades [107]. After a few years, the recombinant acellular pertussis vaccine was developed [111]. It replaced the much more reactogenic former whole-cell pertussis vaccine. Although safer, acellular pertussis seems to be less protective than the other. This may be due to the different immunological components stimulated by the 2 vaccines. The former, alum-adjuvanted, able to preferentially stimulate Th2 (poorly effective in defense) and Th17, but scarcely Th1, and the latter able to strongly stimulate Th1 and Th17, both effective in protection [112]. Two recombinant vaccines were developed for Lyme disease, both composed of the Borrelia burgdorferi outer surface protein A. The first was licensed in 1998, whereas the other was never licensed. As a consequence, in fact, of public concern about adverse events (fear of a never demonstrated vaccine-induced arthritis) and other considerations, including efficacy of approximately 80%, lack of available safety and immunogenicity data for 50-year-old people, and safety comparable to placebo, excepting the solicited local and systemic adverse reactions, which were higher in the vaccine group [141]. In 1994, a very effective, killed HA vaccine, based on the pioneering studies of Hilleman et al. [76], was developed and licensed. This vaccine has demonstrated its usefulness mainly in travelers, to whom, isolated or combined with HB vaccine, may be offered [142]. The possibility to move from an animal model to cell culture methods to grow viruses has enabled to develop effective inactivated vaccines for rabies virus from human diploid or chick embryo cells [143], which may be used in both, pre- and postexposure prophylaxis.

Polysaccharide and conjugate vaccines In the seventies of the last century, following a long interval since the pioneering studies on Streptococcus pneumoniae [144], the plain polysaccharide vaccines for Hib [145], Neisseria meningitidis [146] and Salmonella typhi [147] were developed. Capsular polysaccharides, in fact, may directly stimulate B cells to synthesize protective antibodies, without Tcell intervention. T-independence, however, involves: (1) less effective and short-lasting antibody response, principally oligoclonal [148]; (2) poorly protective immune response in < 2year-old infants for limited switching to IgG2 [149], the main defensive IgG subclass against polysaccharide antigens; (3) lack of memory cell induction. Despite undeniable merits in controlling the diseases induced by the above reported microorganisms in the adults, a series of considerations led to the replacement of plain polysaccharide by conjugate vaccines. These include the poor response in infants, who are preferential targets for the majority of the considered diseases, the less effective and short-lasting immune response, but principally the recently demonstrated responsibility in the progressive reduction of the immune repertoire for lack of memory effect. Conjugate vaccines were obtained according to the method set up by Amery already in 1929 [150], examined in depth by Jennings [151] and realized by Hilleman’s group [152] and others [153,154,155,156], since the 80s of the last century until more recently [157,158]. Conjugation, in fact, with a largely known protein antigen, such as diphtheria and tetanus toxoids, not only transforms T-independent into T-dependent antigens, but also stimulates the immune response, through the recruitment of bystander helper T cells [159]. However, the technical problems of conjugation are also related to the right protein/polysaccharide ratio and the number of polysaccharides to be conjugated to protein. This may be a special problem for Streptococcus pneumoniae, which includes more than 90 different serotypes. Recently, a 13-valent conjugate pneumococcal vaccine has been developed and licensed [160]. The high cost due to technical problems of conjugation and the limited protection, especially in the geographical areas characterized by high prevalence of serotypes not included in the vaccine, prompted some researchers to explore alternative routes for developing cheaper and universally protective vaccines. Trials have been done with the surface pneumococcal proteins A and C, but recently the whole cell vaccine, already tried nearly a century ago, has proved cheap, successful and able to mediate protection through specific antibodies and Th17 cells [161,162].

INTERNATIONAL REVIEWS OF IMMUNOLOGY

13

Table . Main human vaccine characteristics with the approximate year of availability and the main vaccine developer(s).

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

Vaccine

Type

Year

Developer(s)references

Variola Rabies Typhoid

Live attenuated Live attenuated Inactivated

  

Cholera Plague Tuberculosis Diphtheria Tetanus Whole-cell Pertussis

Inactivated Inactivated Live attenuated Subunits (toxoid) Subunits (toxoid) Inactivated

     

Yellow Fever Influenza Polio Streptococcus pneumoniae Polio Measles Mumps Rubella Neisseria meningitidis Adenoviruses Haemophilus influenzae type b Typhoid Hepatitis B

Live attenuated Inactivated Inactivated Subunits (polysaccharides) Live attenuated Live attenuated Live attenuated Live attenuated Subunits (polysaccharides) Live attenuated Subunits (polysaccharides)

          

E Jenner [] L Pasteur [] R Pfeiffer, W Kolle [], AE Wright [] J Ferrán [] W Haffkine [] A Calmette, C Guérin [] G Ramon [] G Ramon [] AH Mayer, M Kristensen, E Sörensen [] M Theiler∗, HH Smith [] JE Salk [] JE Salk [] CM McLeod [] AB Sabin [] JF Enders∗ [] MR Hilleman [] MR Hilleman [] EC Gotschlich et al. [] MR Hilleman [] JB Robbins, R Schneerson []

Live attenuated Subunits (s antigen) plasma-derived Subunits (s antigen) recombinant Live attenuated Conjugated protein/polysaccharide Conjugated protein/polysaccharide Conjugated protein/polysaccharide Subunits (toxin) recombinant Subunits recombinant Inactivated Conjugated protein/polysaccharide Live attenuated reassortant Subunits (OspA) recombinant

 

R Germanier, E Füer [] MR Hilleman []

  

P. Valenzuela et al. [], WJ McAleer [] M Takahashi [] R Schneerson et al. []



R Schneerson et al. []



SC Szou et al. []

   

R Rappuoli et al. [] R Kirnebauer et al. [] MR Hilleman [] CK Fairley et al. []

 

HF Clark, et al. [] SmithKline Beecham []

Hepatitis B Varicella Haemophilus influenzae type b Streptococcus pneumoniae Salmonella typhi Vi Acellular Pertussis Papillomavirus Hepatitis A Neisseria meningitidis Rotavirus Lyme disease

∗ Nobel prize winners (JF Enders not for developing measles vaccine, but for the ability to grow polioviruses in cultures).

A list in chronological order of the main developed vaccines is reported in Table 1, whereas the classification of vaccines with the corresponding types of immune response is summarized in Figure 1.

The biological weapons threat In 1999, the Centers for Disease Control and Prevention (CDC) in the United States, prompted by the fear that microorganisms could be used as weapons, convened a meeting in Atlanta, in order to classify the biological agents according to the level of danger [17,163,164]. The agents were classified into three categories, the most dangerous category A, followed by B and C. In the category A, the included microorganisms were represented by Variola major, the etiologic agent of smallpox, Bacillus anthracis, the etiological agent of anthrax, Yersinia

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

14

E. D’AMELIO ET AL.

Figure . Vaccine classification and corresponding types of immune response.

pestis, the etiological agent of plague, Clostridium botulinum (botulinum toxins), the etiological agent of botulism, Francisella tularensis, the etiological agent of tularemia, Filoviruses and Arenaviruses (e.g., Ebola virus, Lassa virus), the etiological agents of viral hemorrhagic fevers. For all these agents, vaccines are either unsatisfactory or lacking, thus further justifying their inclusion among the dreadful category A agents. Considering that the diseases caused by these microorganisms are either eradicated or relatively uncommon, the stimulus to develop safer and more effective vaccines is weak and principally driven by the fear that they could be used in bioterrorism context. However, the progress towards new vaccines have been already reported for smallpox, anthrax, botulinum toxins and plague, whereas for Francisella tularensis and Ebola are reported below. Francisella tularensis is a Gram-negative bacterium with the potential to induce lifethreatening human diseases, including the glandular form, pneumonia and septicemia, and it results infectious for even low inhalant doses. Killed vaccines have been proven ineffective and a live vaccine strain (LVS) exists as investigational new drug (IND) offered to at-risk laboratory personnel. Recently recombinant attenuated derivatives from a virulent category A strain, SCHU S4, were tested in rabbits. Two out of these derivatives showed partial protection (27–36%) against death, a result better than with LSV, thus providing the first demonstration of protection in an animal species other than a rodent [165].

The twenty-first century and future prospects The current century shows the increasing trend to move from an empirical vaccine development to a rational design [166]. Genetic engineering allows the development of a number of applications [31]. They consist of the vectored vaccines, that is the inclusion of genes coding for

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

15

the protein antigens in live attenuated viral or bacterial vectors [167], such as poxviruses, adenoviruses, BCG [31], Salmonella or Listeria. These latter vectors enable to immunize through the mucosal route [9]. Another possible application is represented by the reverse genetics, that is the preparation of DNA complementary (cDNA) to viral negative RNA strands and reconstitution of the viruses through co-infection of cells with those cDNA plasmids [168,169]. Finally, the reverse vaccinology, that is the inclusion of a microbial gene in Escherichia coli, in order to evaluate the possible interest of the coded proteins for vaccine development [170], thus moving the focus of attention from the microorganism to its genome [171]. By this last approach an effective vaccine against Neisseria meningitidis serogroup B has been recently developed [172]. Its capsular polysaccharide is, in fact, poorly recognized by the immune system. This vaccine includes three proteins present in the majority of meningococcal B strains able to stimulate protective response. By comparative genome analysis an effective vaccine against Streptococcus agalactiae, one of the leading causes of childhood invasive infections in USA and Europe and also an emerging threat in the elderly, has been designed [170]. By reverse vaccinology, the acellular pertussis vaccine has also been realized, and vaccines able to replace the conjugate ones, mainly those with several polysaccharides, may perhaps be developed in the future [173]. Synthetic DNA vaccines appear very promising, but they have not yet been licensed in humans, due to their low potency. However, recent technological advances have generated renewed interest for this new type of vaccines also in humans [174]. Systems vaccinology is a modern approach to the study of vaccine immunity, adopted from systems biology, which describes, in an interdisciplinary and systematic way, the complex mutual interactions among the different components in a biological system, thus analyzing it as a whole. Systems vaccinology has already demonstrated its capacity to predict the response to a vaccine even at individual level and to deepen the scientific knowledge of the immune response to vaccine [175]. Systems vaccinology implies different technologies, including modern mass spectrometry and different microarrays (DNA, antibody and pathogen proteome) [176]. Moreover, identifying molecular signatures which are early expression of different immune functions may help to discover new correlates of protection [175]. Thus, systems vaccinology is a very promising study approach, which has already provided convincing evidence of its extreme potential. In the approximately 130 years since Pasteur’s time, although the principle of attenuation/inactivation has generally been successfully applied, the difficulty in developing effective vaccines in different diseases enabled to realize that it does not always work. BCG, lack of effective vaccines for malaria and HIV, the difficulty in developing effective vaccines for RNA viruses, due to their high variability, witness that a deep knowledge of the microorganism biology and the relative correlates of protection are required in order to design effective vaccines [177]. In particular, the need for yearly influenza vaccine administration, as a consequence of drifted annual antigenic modifications, and the lack of vaccines for hepatitis C virus (HCV) and HIV are examples for such complexity. Moreover, the best vaccines are those addressed towards microorganisms which are completely and permanently cleared by the immune response. Conversely, no effective vaccines have been developed towards microorganisms which are barely challenged by the immune system, such as HCV, HIV, respiratory syncytial virus (RSV), cytomegalovirus, Pseudomonas aeruginosa, Staphylococcus aureus and malaria [9]. Furthermore, in some viral infections, the presence of specific antibodies may not result in protection, but, as in the case of dengue, in infection enhancement [178]. However, despite this possible enhancement, very recently a recombinant, vectored (attenuated 17D yellow fever vector), live, tetravalent dengue vaccine has been used in phase 3 clinical trials in Latin America and Asia in 9- to 16-year-old children. It has been demonstrated to be

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

16

E. D’AMELIO ET AL.

safe and effective. Vaccine efficacy versus placebo, after a three-dose immunization schedule, against virologically confirmed dengue cases of 60.8%, hospitalization for dengue of 80.3% and severe dengue of 95.5%, has been demonstrated [179].Dengue is a mosquito-transmitted viral disease, caused by four different subtypes, responsible for an estimated annual 390 million infected subjects, approximately one quarter of whom are symptomatic [180]. HCV is a flavivirus responsible for an estimated chronic infection of 130–150 million patients and annual death of 350,000–500,000 people worldwide [181]. The current efforts for an HCV vaccine are addressed towards the prime-boost model. Priming with a chimpanzee adenovirus and boost with the MVA as vector, expressing a non structural region from a genotype 1b virus with genetically inactivated RNA-dependent RNA-polymerase activity (NS5b), in order to induce specific T-cell response, is under study [182]. Hepatitis E virus induces a generally self-limiting disease transmitted through fecal-oral route, with a mortality of 1–3%, which may even reach 25% in pregnant women. It is responsible for an estimated worldwide annual 20 million infections, over 3 million of which acute, and an estimated over 56,000 deaths [183]. A recombinant vaccine has been developed and licensed in China and recently it has been demonstrated to induce specific antibodies and protection lasting up to 4.5 years [184]. The history of the efforts spent in the search for an effective HIV vaccine is very instructive as expression of the difficulties encountered in case of a very variable microorganism, able to escape the immune system reaction. In particular, the first trials to develop vaccines including the gp 120 Env protein have been unsuccessful [185,186], as well as the adenovirus vectored vaccine containing clade B Gag, Pol and Nef, without Env, with the intent to activate cellular immune response. Actually, despite the appearance of specific anti-Pol and Nef CD8+ T-cell response in the majority of vaccinees, viral levels were not decreased [187,188,189]. More recently, a phase III study in Thailand of 2 HIV vaccines in combination, one canarypox vector encoding Gag, Env and protease for priming and one recombinant subunit Env gp 120 for boosting (prime-boost strategy), has achieved 31.2% of protection. Although low, it is the first demonstration that protection from HIV may be vaccine-induced. Furthermore, the correlates of protection have been identified in antibodies specific for V1 and V2 variable regions of gp 120 [190,191]. Currently, biotechnological approaches may help identifying invariant relevant epitopes even in RNA hyper-variable viruses, such as HIV and influenza, through the use of broadly neutralizing monoclonal antibodies [192,193,194,195]. The rational design of universal influenza and HIV vaccines may, therefore, be facilitated. Moreover, the conceptual design of innovative vaccines may also take advantage of and be driven by deep sequencing. That is the capacity to generate millions of sequence of the immunoglobulin genes, to identify intermediates to maturation of the broadly neutralizing antibodies [195,196,197]. RSV is responsible for the induction of low respiratory infections in approximately 30 million children annually, 10% of whom require hospitalization, and of approximately 200,000 deaths, generally in < 5-year-old children. RSV may interfere with the host immune system, by stimulating inflammation and a Th2 cell population able to dampen cytotoxic T cell activity and viral clearance. Despite efforts for developing a vaccine date back to the sixties of the last century, no vaccine has been approved and licensed yet. This may at least partly be linked to the unsuccessful formalin-inactivated RSV (FI-RSV) vaccine, which was able to stimulate a florid, non-protective, antibody response. Rather, the disease with wild-type RSV was more severe than in non-vaccinated subjects, and two vaccinated children died with a clinical picture of lower respiratory infection and the presence of RSV in the lungs [198]. The reasons for such behavior are not completely understood; it has been hypothesized that formalin was

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

17

able to inactivate protective epitopes, thus stimulating the synthesis of non-protective, nonneutralizing, antibodies, and even failing to induce protective CD8+, RSV-specific, cytotoxic T-lymphocytes [199]. FI-RSV vaccine failure witnesses the difficulty of vaccine development, if the virus biology is not completely known and the approach to vaccine preparation is largely empirical. The majority of vaccines currently under study are at preclinical phase and only two attenuated vaccines have been successfully tested in infants through intranasal route [200]. Cytomegalovirus is a herpesvirus, which represents the most frequent cause of congenital birth defect, with an estimated 0.6% annual incidence. Moreover it may cause severe diseases in immune- compromised individuals, such as transplant recipients. The search for a safe and effective vaccine is, therefore, of pivotal relevance. Recently, by a method denominated “analytic vaccinology”, consisting in the careful analysis of the specific anti-cytomegalovirus antibodies, in order to identify the neutralizing epitope(s), it has been possible to identify a highly immunogenic pentamer. It was capable, in fact, to elicit, when administered to mice in Chinese hamster ovary (CHO) cells, powerful neutralizing antibody response, at a titer 1,000-fold higher than those found in the sera of convalescent donors [201]. Pseudomonas aeruginosa is a gram-negative bacterium, particularly resistant to antibiotics, able to induce life-threatening infections in immune-compromised patients, thus making the search for an effective vaccine urgent. Several P. aeruginosa antigens have been tested as candidate vaccines, including lipopolysaccharide O antigens, flagella, outer membrane proteins, and antigens of the type III secretion system, but they, although protective and promising, have not yet provided conclusive results [202]. Staphylococcus aureus is a gram-positive microorganism, causing life-threatening infections in humans, equipped with several virulence factors and able to evade the defensive activity of the immune system. Moreover, it is resistant to the majority of the antibiotics. This makes the search for a vaccine necessary but difficult, for a series of reasons, including the need for vaccine to contain different virulence factors, which are often poorly known, and the lack of knowledge of the correlates for protection. However, very recently a candidate combination vaccine, including five different virulence factors in its composition, mixed with a new alum adjuvant linked to a Toll-like receptor 7 agonist, was able to protect nearly 100% of mice challenged with four different staphylococcal strains. Protection was associated to high specific antibody titers and Th1 skewed immune response. Moreover, low frequencies of Th17 cells were also present [203]. Malaria presents a global annual incidence of approximately 200 million people and 1.2 million deaths [204]. Recently, after decades of unsuccessful trials, a phase III trial of a recombinant vaccine containing 2 antigens, one circumsporozoite protein and the surface antigen of HB virus, with the adjuvant AS01, showed 55% of protection in 5- to 17-month age group, but lower (34.8%) in 6- to 12-week-old infants [205]. Despite incomplete protection, this vaccine seems to be the first reliable malaria vaccine candidate to move forward. Ebola hemorrhagic fever is a high mortality, dreadful disease, induced by one filovirus, first described in 1976 in Africa, in a border area between south of Sudan and north of the former Zaire, and episodically reappearing in self-limiting outbreaks. Ebola virus, together with other filoviruses and arenaviruses, like Marbourg and Lassa, has been included among the dreadful category A biological agents. The last Ebola epidemic in West Africa, which has started in the first half of 2014 and is still ongoing, at the end of May 2015 has registered over 27,000 infected patients, over 11,000 (41%) of whom died. A candidate vaccine, formed by the vesicular stomatitis virus (rVSV) as vector, genetically modified to express Zaire strain Ebola virus glycoproteins (rVSV-ZEBOV), developed by NewLink Genetics and Merck Vaccines

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

18

E. D’AMELIO ET AL.

USA in collaboration with the Canadian Public Health Agency, after having successfully concluded a Phase I clinical trial last January [205], is currently in phase II and phase III clinical trials in Guinea and Sierra Leone. The strategy will be ring vaccination, that is the vaccination of contacts of confirmed, probable and suspected cases, the same strategy used for the successful smallpox eradication. Moreover, the vaccine will be offered to frontline health care workers. The interim analysis has shown that rVSV-ZEBOV might be highly effective and safe in preventing Ebola [206]. Another vaccine, the chimpanzee adenovirus vectored recombinant vaccine (chAd3-ZEBOV), developed by GlaxoSmithKline in collaboration with the US National Institute of Allergy and Infectious Diseases, has successfully completed phase I clinical trial last January, is performing phase II clinical trial and planning phase III. Johnson & Johnson, in association with Bavarian Nordic, has developed a heterologous prime-boost vaccine, known as Ad26-EBOV and MVA-EBOV, currently in phase I clinical trial. Novavax, a biotech company in the USA, is developing a recombinant Ebola vaccine based on Guinea 2014 Ebola strain. Finally, Russian Federation is developing different Ebola candidate vaccines, some of which vectored with influenza virus (phase I trial will start in the second half of 2015), or adenovirus or VSV or recombinant rabies virus [207]. Clostridium difficile is annually responsible in the USA alone of an estimated 500,000 infections and 15,000–20,000 deaths with an annual cost of approximately $ 3.2 billion [208]. Considering that this infection is on the increase and that hyper-virulent strains are emerging, the problem to develop protective vaccines has been faced since a few years. Despite many candidates have been identified, some perplexity still remains on the optimal vaccine composition, in order to completely detoxify the toxins and act not only for improving symptoms but also for avoiding colonization [209].

The adjuvants The more the antigens are purified the less they are immunogenic, thus they need to be associated with an adjuvant, that is a substance capable of increasing the immune response. The term “adjuvant” comes from the latin adjuvare, which means “to help.” It was used for the first time by Ramon to denominate the substances (tapioca, saponin, agar, etc.) used to increase the immune response to diphtheria and tetanus toxoids [210]. For more than 70 years, the only approved adjuvant for human use was represented by aluminum phosphate or hydroxide (alum), which works by stimulating inflammasome, Th2 and humoral immune response, but poorly Th1 [211]. More recently, other adjuvants, single or alum-associated, have been approved, able to more strongly stimulate antibody response and induce a shift from Th2 to Th1 [212]. Among the different candidates for alum substitution, phospholipid bilayer vesicles (liposomes) are very promising. Virosomes (liposomes composed by influenza membrane lipids and glycoproteins, supplemented with phosphatidylcholine [PC]) are included in licensed influenza and hepatitis A vaccines since 1997 [213]. Moreover, MF59, an oil-in-water emulsion, including a low percentage (4.3%) of biodegradable squalene oil, stabilized with Tween 80 and Span 85 in citrate buffer from Novartis, incorporated in influenza vaccine, has been licensed and safely employed in million subjects. Even AS04, 3–0-desacyl-4 -monophosphoryl lipid A (MPL) derivative from lipopolysaccharide (LPS) of Salmonella Minnesota detoxified and linked with alum, from GSK Biologics, has been licensed and incorporated in HB and papillomavirus vaccines [214]. MF59 works very well, by increasing level, diversity and affinity of antibody response to influenza pandemic vaccine [215,216]. Despite the reported lack of effectiveness

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

19

Figure . Chronology of licensed adjuvants, adjuvanted vaccines and their main mechanisms of action.

of MF59 if simultaneously, but separately, administered with vaccine [217], actually it was able to stimulate the antibody response to a non-adjuvanted, seasonal influenza vaccine, when the adjuvanted pandemic vaccine was simultaneously, but separately, administered [218]. AS03 is an adjuvant very similar to MF59, licensed for influenza vaccines by GSK Biologics. The AS03adjuvanted pandemic influenza vaccine has been associated with the appearance of cases of narcolepsy after vaccination in some North European Countries [219], whereas this has not been observed with the MF59-adjuvanted pandemic flu vaccine [220]. This enables to infer that such differential behavior may be linked to the α-tocopherol, which is included in the AS03, but not in the MF59, composition [221]. Quite different, instead, is AS04, which works as an agonist of Toll-like receptor 4, thus stimulating Th1 cells and complement-fixing antibodies [222]. The chronology of these licensed adjuvants, the adjuvanted vaccines and their main mechanisms of action [223.224,225,226,227,228,229] are summarized in Figure 2. Finally, AS02, which is used in the attempts to develop vaccines against HIV, tuberculosis and malaria, is an oil-in-water emulsion including QS21 (an extract of the bark of the South American tree Quillaja saponaria) and MPL, whereas AS01 contains also liposomes [214]. However, all the approved adjuvants, including alum, the newer emulsions and LPS derivative with alum, act by primarily stimulating innate immunity, which in turn activates specific immune response. This behavior mimics the response to natural infections and live viral

20

E. D’AMELIO ET AL.

Table . Global infant EPI vaccine coverage in . Vaccine

Global coverage

DTP BCG Measles HBV Polio

% % % >% %

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

DTP = Diphtheria,/Tetanus/Pertussis, third dose; BCG = Bacille Calmette-Guérin; HBV = Hepatitis B virus.

vaccines, such as yellow fever and influenza, where the gene signatures of innate anti-viral activation are precociously evident, as explored by systems vaccinology [56,230].

Special people categories needing tailored vaccination programs In order to take full advantage of the vaccine potential, the highest possible global vaccine coverage should be promoted. Routine infant immunization should be further expanded. Regarding the EPI-included vaccines, in fact, the global 2013 coverage may be estimated in 84% for the third dose of the trivalent DTP, 90% for BCG, 84% for measles, > 94% for HB and 84% for polio, with only 3 remaining wild poliovirus endemic countries (Table 2), thus allowing to register approximately 13% of children not yet covered [231]. Moreover, the promotion of systematic vaccination of selected people categories according to the specific risk profile should be actively pursued. Regarding the travelers, WHO and CDC recommend to be up-to-dated with routine immunization, including MMR, DTP, and polio, varicella and influenza (CDC only). Moreover, yellow fever (compulsory for travels to some endemic countries), HA (strongly recommended even though administered the same day of departure [232]), HB, meningococcal meningitis, Japanese encephalitis, rabies, typhoid, and cholera vaccines, depending on destination and in special circumstances, are even recommended. Regarding the military, vaccination programs are indeed already present in 90% of countries [233], frequently introduced well before, even decades, and exceeding, the vaccination programs for civilian population [234] and generally rightly implemented [233]. This underlines the role of the military in vaccinology, with military researchers having invented, developed or improved at least 20 vaccinations [234]. Moreover, especially in the countries where military service is compulsory, screening for infectious diseases and vaccination programs substantially contribute to the fight against infectious diseases, thus complementing the activity of the civilian health services [233]. In the pregnant women living vaccines should be avoided during pregnancy, whereas no limitation is considered with inactivated or subunit vaccines [235], as influenza, tetanus and pertussis, which are recommended and result protective for the fetus and newborn too [9]. Healthcare workers, who may become source of vaccine-preventable infections [236], should at least be immunized with influenza and HB vaccines. Moreover, they should maintain updated high coverage for all the vaccine-preventable diseases endemic for the country and according to the occupational risk [237]. The food handlers should receive typhoid [238] and HA [239] vaccines. Patients with chronic diseases on immunosuppressive therapy, who are particularly exposed to infections [240] and generally not adequately covered [241,242], and their household and close contacts should at least receive influenza and pneumococcal vaccinations. They

INTERNATIONAL REVIEWS OF IMMUNOLOGY

21

Table . Special people categories needing tailored vaccination programs.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

• Travelers • Military • Pregnant women • Healthcare workers • Food handlers • Patients with chronic diseases on immunosuppressive therapy and their household & close contacts • Elderly • Potential targets of bioterrorism threats

are, in fact, strongly recommended by the European League against Rheumatism (EULAR) [243], but scarcely administered worldwide [244]. Moreover HB vaccine should be offered to seronegative patients [245,246], whereas vaccines for papillomavirus and for shingles (which is currently a live attenuated vaccine) should be evaluated according to the specific risk of infection. The elderly should receive influenza, conjugate pneumococcal and shingles vaccines [247]. The potential targets of bioterrorism threats should receive vaccines for unusual pathogens, such as CDC category A, [17,248], or prophylaxis according to the specific threats (Table 3).

Conclusions The history of vaccination is strictly and reciprocally interwoven with the development of microbiology, immunology and molecular biology. Effective vaccines have been developed by applying advances in these fields, and, conversely, the study of successful vaccines may promote advances in immunology [149]. It has been estimated that vaccines annually prevent almost 6 million deaths worldwide [249] and that in the United States a 99% reduction of the nine diseases for which specific vaccinations were recommended for decades has been registered [2]. Moreover, the annual return of investment for vaccination has been calculated to be in the range of 12–18%, even though there may be a large underestimation [1]. Finally, the vaccine effect is also addressed to the prevention of consequences of infectious diseases, including cancer, as in the case of HB and papillomavirus vaccines. Much work should still be done for improving old, but unsatisfactory, vaccines, such as BCG [250], anthrax [23], smallpox [251], plague [252], or for developing effective vaccines for HIV [253], malaria [205], HCV [182], dengue [254], RSV [255], cytomegalovirus [256], multi-resistant bacteria [203], Clostridium difficile [208,209] and Ebola virus [257] (Table 4). Moreover, many efforts are also addressed to find more and more immunogenic, safe, and easy to use (in relation to route of administration and number of contemporaneously administered antigens) vaccine preparations. Recently, an aerosolized measles vaccine has demonstrated to be immunogenic, but inferior to the subcutaneous vaccine with respect to the rate of seropositivity [258]. The continuous reduction of companies able to produce vaccines, that may cause concern on the capacity to respond, should an outbreak occur, and the high cost for developing a vaccine, which may be calculated in $ 500–1,000 million [259], make the research for innovative vaccines difficult. Moreover, these conditions may also make the regular vaccine supply to developing countries not sure, despite the international efforts, through WHO, United Nations Children’s Fund (UNICEF) and the Foundations, such as the Bill and Melinda Gates Foundation [31]. Economic and organizational reasons (cold chain maintenance, need for the presence of healthcare workers for parenteral administration, etc.) are the main constraints for global full vaccine coverage in developing countries. However, vaccines are even underused in affluent

22

E. D’AMELIO ET AL.

Table . Prospects for future vaccines.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

Infectious disease/microorganism Tuberculosis Anthrax Smallpox Plague HIV Malaria Hepatitis C virus Dengue Respiratory syncytial virus Cytomegalovirus Multiresistant bacteria (Staphylococcal) Clostridium difficile Ebola virus

Vaccine has to be

Type of vaccine references

Improved Improved Improved Improved Developed Developed Developed Developed Developed Developed Developed

Live Recombinant/Vectored vaccine [] Recombinant [] Attenuated/Subunit/Recombinant [] Live/Recombinant subunit [] Vectored + Recombinant subunit [] Recombinant [] Vectored prime-boost (adenoviral+MVA) [] Recombinant live vectored (YF) [] Attenuated/Mucosal [] Recombinant Pentamer [] Recombinant Combined Virulence Factors []

Developed Developed

Recombinant Toxin Subunits [,] Vectored recombinant (VSV-chAD) []

HIV: human immunodeficiency virus; MVA: Modified Vaccine Ankara; YF: yellow fever; VSV: vesicular stomatitis virus; chAD: chimpanzee adenovirus .

countries, frequently for concern on safety, despite the successes in controlling infectious diseases and the documented safety. A Medline search for “vaccine risks” largely overcome (approximately five-fold) the search for “vaccine benefits” [1]. Recently, the legal and ethical issues linked to the need to protect voluntarily unvaccinated children during outbreaks through social distancing measures has been discussed [260]. Vaccines are administered to healthy people, who are scarcely prone to undergo medical treatment, thus a continual education program should be performed, to make people aware that vaccines represent a precious resource, in order to promote voluntary adhesion. Only a great and coordinated global effort among different societal components (university, research centers, pharmaceutical industry, ministry of health), joined to a continual education addressed to both, healthcare personnel and general population, will allow people be broadly covered.

Declaration of interest The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the article.

References [1] André FE, Booy R, Bock HL, et al. Vaccination greatly reduces disease, disability, death and inequity worldwide. Bull WHO 2008;86:81–160. [2] Anon. Impact of vaccines universally recommended for children. 1900–1998. MMWR 1999;48:243–248. [3] Paran N, Sutter G. Smallpox vaccines. New formulations and revised strategies for vaccination. Hum Vaccin 2009;5:824–831. [4] Henderson DA, Klepac P. Lessons from the eradication of smallpox: an interview with D. A. Henderson. Philos Trans R Soc Lond B Biol Sci 2013;368:20130113. [5] Chia WN, Goh YS, Rénia L. Novel approaches to identify protective malaria vaccine candidates. Front Microbiol. 2014 Nov 17;5:586. [6] Karp CL, Wilson CB, Stuart LM. Tuberculosis vaccines: barriers and prospects on the quest for a transformative tool. Immunol Rev. 2015;264:363–381. [7] John M, Gaudieri S. Influence of HIV and HCV on T cell antigen presentation and challenges in the development of vaccines. Front Microbiol. 2014;5:514.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

23

[8] Dunning R. Some observations on vaccination or the inoculated Cow-Pox. London: March and Teape; 1800. p. 5. [9] Delany I, Rappuoli R, De Gregorio E. Vaccines for the 21st century. Embo Mol Med 2014;pii:emmm.201403876v1. doi: 10.1002/emmm.201403876 [10] Pulendran B, Ahmed R. Immunological mechanisms of vaccination. Nat Med 2011;12:509–517. [11] Jenner E. An enquiry into the causes and effects of variolae vaccinae a disease discovered in some western countries of England, particularly Gloucestershire, and known by the name of Cow Pox. London: printed for the author by Sampson Low; 1798. [12] Cha SH. The history of vaccination and current vaccination policies in Korea. Clin Exp Vaccine Res 2012;1:3–8. [13] Baxby D. Edward Jenner’s role in the introduction of smallpox vaccine. In: Stanley Plotkin, editor. History of vaccine development. New York: Springer Science+Business Media, LLC; 2011. pp. 13–19 [14] Behbehani AM. The smallpox story: life and death of an old disease. Microbiol Rev 1983;47:455– 509. [15] Riedel S. Edward Jenner and the history of smallpox and vaccination. Proc (Bayl Univ Med Cent) 2005;18:21–5. [16] Grabenstein JD, Pittman PR, Greenwood JT, et al. Immunization to protect the US Armed Forces: heritage, current practice, and prospects. Epidemiol Rev. 2006;28:3–26. [17] Rotz LD, Khan AS, Lillibridge SR, et al. Public health assessment of potential biological terrorism agents. Emerg Infect Dis. 2002;8:225–230. [18] Artenstein AW, Grabenstein JD. Smallpox vaccines for biodefense: need and visibility. Expert Rev. Vaccines 2008;7:1225–1237. [19] Berche P. Louis Pasteur: from crystals of life to vaccination. Clin Microbiol Infect 2012;18(Suppl.5):1–6. [20] Pasteur L. Sur les maladies virulentes et en particulier sur la maladie appelée vulgairement choléra des poules. CR Acad Sci (Paris) 1880; 90:239–248. [21] Pasteur L. De l’atténuation du virus du cholera des poules. CR Acad Sci (Paris) 1880; 91:673–80. [22] Pasteur L, Chamberland CE. Sur la vaccination charbonneuse. CR Acad Sci (Paris) 1881;92:1378–1383. [23] Kaur M, Singh S, Bhatnagar R. Anthrax vaccines: present status and future prospects. Expert Rev Vaccines. 2013;12:955–970. [24] Pasteur L. Méthode pour prévenir la rage aprés morsure. C R Acad Sci (Paris) 1885;101:765–774. [25] Succi RC, Farhat CK. Vaccination in special situations. J Pediatr (Rio J) 2006;82:S91–100. [26] Salmon DE, Smith T. On a new method of producing immunity from infectious diseases. Am Vet Rev 1886;10:63–69. [27] Pfeiffer R, Kolle W. Experimental investigation of prevention of typhoid fever in man by immunization. Dtsch. Med. Wochenschr 1896;22:735–737. [28] Wright AE, On the results which have been obtained by anti-typhoid inoculation. Lancet 1902;2:651–654. [29] Bornside GH. Waldemar Haffkine’s cholera vaccines and the Ferran–Haffkine priority dispute. J Hist Med All Sci 1982;37:399–422. [30] Haffkine WM. Remarks on the plague prophylactic fluid. Br Med J 1897;1:1461–1462. [31] Plotkin SA. Vaccines: past, present and future. Nat Med Suppl 2005;11:S5-S11. [32] Jefferson T, Demicheli V, Pratt M. Vaccines for preventing plague. Cochrane Database Syst Rev 2000;(2):CD000976. [33] Williamson ED, Oyston PCF. Protecting against plague: towards a next-generation vaccine. Clin Exp Immunol 2012;172:1–8. [34] Tuells J. Vaccinology: the name, the concept, the adjectives. Vaccine 2012;30:5491–5495. [35] Tan SY, Kwok E. Albert calmette (1863–1933): originator of the BCG vaccine. Singapore Med J 2012;53:433–434. [36] Koch R. The current state of the struggle against tuberculosis. Nobel lecture December 12 1905. From nobel lectures. Physiology or Medicine 1901–1921. Amsterdam: Elsevier Publishing Company; 1967.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

24

E. D’AMELIO ET AL.

[37] Calmette A, Guérin C, Breton M. Contribution a l’étude de la tuberculose expérimental du cobaye (infection et essays de vaccination par la voie digestive). Ann Inst Pasteur Paris 1907;21: 401–416. [38] Luca S, Mihaescu T. History of BCG Vaccine. MAEDICA J Clin Med 2013;8:53–58. [39] Clemens JD. The BCG controversy: a methodological and statistical reappraisal. JAMA 1983;249:2362–2369. [40] Mangtani P, Abubakar I, Ariti C, et al. Protection by BCG vaccine against tuberculosis: a systematic review of randomized controlled trials. Clin Infect Dis. 2014;58:470–480. [41] Bourdin Trunz B, Fine P, Dye C. Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of cost-effectiveness. Lancet 2006;367:1173–1180. [42] Rodrigues LC, Mangtani P, Abubakar I. How does the level of BCG vaccine protection against tuberculosis fall over time? BMJ 2011;343:d5974. [43] Jensen K, Ranganathan UD, Van Rompay KK, et al. A recombinant attenuated Mycobacterium tuberculosis vaccine strain is safe in immunosuppressed simian immunodeficiency virusinfected infant macaques. Clin Vaccine Immunol. 2012;19:1170–1181. [44] Hesseling AC, Johnson LF, Jaspan H, et al. Disseminated bacille Calmette-Guérin disease in HIVinfected South African infants. Bull. World Health Organ. 2009;87:505–511. [45] Roth AE, Stensballe LG, Garly ML, et al. Beneficial non-targeted effects of BCG: ethical implications for the coming introduction of new TB vaccines. Tuberculosis 2006;86:397–403. [46] Ritz N, Mui M, Balloch A, et al. Nonspecific effect of Bacille Calmette–Guérin vaccine on routine immunisations. Vaccine 2013;31:3098–3103. [47] World Health Organization—Tuberculosis: a global emergency. Geneva, Switzerland: WHO; 1994. [48] Lawn SD, Zumla AI Diagnosis of extrapulmonary tuberculosis using the Xpert((R)) MTB/RIF assay. Expert Rev Anti-Infect Ther 2012;10:631–635. [49] Theiler M, Smith HH. The use of yellow fever virus by in vitro cultivation for human immunization. J Exp Med 1937;65;787–800. [50] Frierson JG. The Yellow Fever Vaccine: a History. Yale J Biol Med 2010;83:77–85. [51] Sellards W, Mathis C, Laigret J. Sensibilité du Macacus rhesus au virus de la fièvre jaune. CR Acad Sci. 1928;186:604–606. [52] Theiler M, Smith HH. The effect of prolonged cultivation in vitro upon the pathogenicity of yellow fever virus. J Exp Med. 1937;65:767–786. [53] Monath TP. Yellow fever vaccines: the success of empiricism, pitfalls of application, and transition to molecular vaccinology. In: Plotkin SA, editor, History of vaccine development. New York: Springer; 2011. p. 119. [54] Available at http://www.who.int/vaccines/en/yellowfever.shtml. [55] Querec T, Bennouna S, Alkan S, et al. Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity. J Exp Med. 2006;203: 413– 424. [56] Querec TD, Akondy RS, Lee EK, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol. 2009; 10:116–125. [57] Gaucher D, Therrien R, Kettaf N, et al. Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J Exp Med. 2008;205:3119–3131. [58] Roux E, Yersin A. Contribution à l’étude de la diphtérie. Ann Inst Pasteur 1888;2:421–499. [59] Faber K. Die Pathogenie des Tetanus. Berl Klin Woch 1890;27:717–720. [60] Linton DS. Emil Von Behring. Infectious Diseases, Immunology, Serum Therapy. Philadelphia: American Philosophical Society; 2005. [61] Ramon G. Sur le pouvoir floculant et sur le proprietés immunisantes d’une toxine diphterique rendu anatoxic (anatoxine). CR Acad Sci (Paris) 1923;177:1338–1340. [62] Ramon G, Zeller C. De la valeur antigenique de l’anatoxine tétanique chez l’homme. CR Acad Sci (Paris) 1926;182:245–247. [63] Smith LA. Botulism and vaccines for its prevention. Vaccine 2009;27:D33–D39. [64] Meyer AH, Kristensen M, Sörensen E. Whooping-Cough vaccination. Acta paediat. 1924;4:21. [65] Anon. Pertussis vaccines: WHO position paper. Wkly Epidemiol Rec 2010;85:385–400.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

25

[66] WHO. Immunization, vaccines and biologicals. Pertussis 2013 global figures. Available at www.who.int/immunization/monitoring_surveillance/Burden/vpd [67] Available at http://www.cdc.gov/pertussis/surv-reporting/cases-by-year.htm. [68] Rota MC, Ausiello CM, D’Amelio R, et al. Prevalence of markers of exposure to Bordetella pertussis among Italian young adults. Clin Infect Dis. 1998;26:297–302. [69] Smith W. The complement-fixation reaction in influenza. Lancet 1936;2:1256–1259. [70] Chenoweth A, Waltz AD, Stokes Jr J, et al. Active immunization with the viruses of human and swine influenza. Am J Dis Children 1936;52:757–758. [71] Francis T Jr, Magil TP. The antibody response of human subjects vaccinated with the virus of human influenza. J Exp Med 1937;65:251–259. [72] Salk JE, Lavin GI, Francis T. The antigenic potency of epidemic influenza virus following inactivation by ultraviolet radiation. J Exp Med 1940;72:729–745. [73] Nakaya HI, Wrammert J, Lee EK, et al. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol. 2011;12:786–779. [74] Phillips CJ, Woolpert T, Sevick C, et al. Comparison of the effectiveness of trivalent inactivated influenza vaccine and live, attenuated influenza vaccine in preventing influenza-like illness among US military service members, 2006–2009. Clin Infect Dis. 2013;56:11–19. [75] Lee YT, Kim KH, Ko EJ, et al. New vaccines against influenza virus. Clin Exp Vaccine Res. 2014;3:12–28. [76] Hoft DF, Babusis E, Worku S, et al. Live and inactivated influenza vaccines induce similar humoral responses, but only live vaccines induce diverse T-cell responses in young children. J Infect Dis. 2011;204:845–853. [77] Lu Y, Welsh JP, Swartz JR. Production and stabilization of the trimeric influenza hemagglutinin stem domain for potentially broadly protective influenza vaccines. Proc Natl Acad Sci USA. 2014;111:125–130. [78] Weller TH, Robbins FC, Enders JF. Cultivation of poliomyelitis virus in cultures of human foreskin and embryonic tissues. Proc Soc Exp Biol Med 1949;72:153–155. [79] Hilleman MR. Vaccines in historic evolution and perspective: a narrative of vaccine discoveries. Vaccine 2000;18:1436–1447. [80] Yun SI, Lee YM. Japanese encephalitis: the virus and vaccines. Hum Vaccin Immunother. 2014;10:263–267. [81] Centers for Disease Control and Prevention. Epidemiology and prevention of vaccinepreventable diseases. Poliomyelitis. Atkinson W, Wolfe S, Hamborsky J, McIntyre L, editors, 11th ed. Washington DC: Public Health Foundation; 2015; pp. 297–310. [82] Howe HA, Bodian D. Poliomyelitis in the chimpanzee: a clinical pathological study. Bull Johns Hopk Hosp 1941;69:149–181. [83] Bodian D, Morgan IM, Howe HA. Differentiation of types of poliomyelitis viruses; the grouping of fourteen strains into three basic immunologic types. Am J Hyg 1949;49:234–245. [84] Melnick JL, Horstmann DM. Active immunity to poliomyelitis in chimpanzees following subclinical infection. J Exp Med 1947;85:287–303. [85] von Magnus H, Melnick JL. Antibody response in monkeys following oral administration of poliomyelitis virus. J Immunol 1948;60:583–596. [86] Bodian D, Horstman DM. Polioviruses. In: Horsfall FL, Tamm I, editors, Viral and Rickettsial Infection of Man, 4th ed, Philadelphia: Lippincott; 1965, p. 430–473. [87] Salk JE. Poliomyelitis vaccine preparation and administration; analysis of basic premises and current practices. JAMA 1959;169:1829–1838. [88] Monto AS. Francis field trial of inactivated poliomyelitis vaccine: background and lessons for today. Epidemiol Rev 1999;21:7–23. [89] Strebel PM, Sutter RW, Cochi SL, et al. Epidemiology of poliomyelitis in the United States one decade after the last reported case of indigenous wild virus-associated disease. Clin Infect Dis 1992;14:568–579. [90] Baicus A. History of polio vaccination. World J Virol 2012;1:108–114. [91] Koprowski HL, Jervis GA, Norton TW. Immune responses in human volunteers upon oral administration of a rodent-adapted strain of poliomyelitis. Am J Hyg 1952;55:108–1126. [92] Melnick J, Plotkin S. Oral polio vaccine and the results of its use. In: Stanley Plotkin, editor. History of vaccine development. New York: Springer; 2011. p. 170.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

26

E. D’AMELIO ET AL.

[93] Sabin AB, Ramos-Alvarez M, Alvarez-Amezquita J, et al. Live, orally given poliovirus vaccine. Effects of rapid mass immunization on population under conditions of massive enteric infection with other viruses. JAMA 1960;173:1521–1526. [94] Mundel T, Orenstein WA. No country is safe without global eradication of poliomyelitis. N Engl J Med 2013;369:2045–2046. [95] Hoke CH Jr, Snyder CE Jr. History of the restoration of adenovirus type 4 and type 7 vaccine, live oral (Adenovirus Vaccine) in the context of the Department of Defense acquisition system. Vaccine. 2013;31:1623–1632. [96] Enders JF, Katz SL, Milovanovic MV, et al. Studies on an attenuated measles-virus vaccine. I. Development and preparations of the vaccine: techniques for assay of effects of vaccination. N Engl J Med 1960;263:153–159. [97] Buynak EB, Hilleman MR. Live attenuated mumps virus vaccine 1. Vaccine development. Proc Soc Exp Biol Med 1966;123:768–775. [98] Hilleman MR, Buynak EB, Weibel RE, et al. Current concepts: live attenuated rubella-virus vaccine. New Engl J Med 1968;279:300–303. [99] D’Amelio R, Biselli R, Fascia G, et al. Measles-mumps-rubella vaccine in the Italian armed forces. JAMA 2000;284:2059. [100] Lista F, Faggioni G, Peragallo MS, et al. Molecular analysis of early postvaccine mumps-like disease in Italian military recruits. JAMA 2002;287:1114–1115. [101] Anon. Global control and regional elimination of measles, 2010–2011. MMWR 2013;62:27–31. [102] Loucq C. Vaccines today, vaccines tomorrow: a perspective. Clin Exp Vaccine Res 2013;2:4–7. [103] Wakefield AJ, Murch SH, Anthony A, et al. Ileal-lymphoid-nodular hyperplasia, non-specific colitis, and pervasive developmental disorder in children. Lancet 1998;351:637–641. [104] Retraction–Ileal-lymphoid-nodular hyperplasia, non-specific colitis, and pervasive developmental disorder in children. Lancet 2010;375:445. [105] Blumberg BS, Alter H. A “new” antigen in leukemia sera. JAMA 1965;191:101–106. [106] Prince AM. An antigen detected in the blood during the incubation period of serum hepatitis. Proc Natl Acad Sci USA 1968;60:814–821. [107] WHO. Hepatitis B. Fact sheet N°204, updated March 2015. Available at www.who.int/mediacentre/ factsheets/fs204/en/ [108] Valenzuela P, Medina A, Rutter WJ, et al. Synthesis and assembly of hepatitis B virus surface antigen particles in yeast. Nature 1982;298:347–350. [109] McAleer WJ, Buynak EB, Maigetter RZ, et al. Human hepatitis B vaccine from recombinant yeast. Nature 1984;307:178–180. [110] Hilleman MR, Buynak EB, Roehm RR, et al. Purified and inactivated human hepatitis B vaccine: progress report. Am J Med Sci 1975;270:401–404. [111] Rappuoli R, Pizza M, De Magistris MT, et al. Development and clinical testing of an acellular pertussis vaccine containing genetically detoxified pertussis toxin. Immunobiology 1992;184:230– 239. [112] Sutton CE, Higgins S, Allen AC, et al. Relative Contribution of Th1 and Th17 Cells in Adaptive Immunity to Bordetella pertussis: Towards the Rational Design of an Improved Acellular Pertussis Vaccine. PLoS Pathog 2013;9:e1003264. [113] Poland GA. Vaccines against Lyme disease: What happened and what lessons can we learn? Clin Infect Dis. 2011;52(Suppl 3):s253–s258. [114] Müller M, Gissmann L. A long way: history of the prophylactic papillomavirus vaccine. Dis Markers. 2007;23:331–336. [115] Centers for Disease Control and Prevention (CDC). FDA licensure of quadrivalent human papillomavirus vaccine (HPV4, Gardasil) for use in males and guidance from the Advisory Committee on Immunization Practices (ACIP). MMWR 2010;59:630–632. [116] Zhou J, Sun XY, Stenzel DJ, et al. Expression of vaccinia recombinant HPV 16 L1 and L2 ORF proteins in epithelial cells is sufficient for assembly of HPV virion-like particles. Virology 1991;185:251–257. [117] Ghim SJ, Jenson AB, Schlegel R. HPV-1 L1 protein expressed in cos cells displays conformational epitopes found on intact virions. Virology 1992;190:548–552. [118] Rose RC, Bonnez W, Reichman RC, et al. Expression of human papillomavirus type 11 L1 protein in insect cells: in vivo and in vitro assembly of viruslike particles. J Virol. 1993;67:1936–1944.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

27

[119] Kirnbauer R, Taub J, Greenstone H, et al. Efficient self-assembly of human papillomavirus type 16 L1 and L1-L2 into virus-like particles. J Virol. 1993;67:6929–6936. [120] Plotkin S. History of vaccination. Proc Natl Acad Sci USA 2014;111:12283–12287. [121] Joura EA, Giuliano AR, Iversen OE, et al. Broad Spectrum HPV Vaccine Study. A 9-valent HPV vaccine against infection and intraepithelial neoplasia in women. N Engl J Med. 2015;372:711– 723. [122] Parashar UM, Hummelman EG, Bresee JS, et al. Global illness and deaths caused by rotavirus disease in children. Emerg Infect Dis 2003;9:565–571. [123] Ward RL, Bernstein DI, Smith VE, et al. Rotavirus immunoglobulin A responses stimulated by each of 3 doses of a quadrivalent human/bovine reassortant rotavirus vaccine. J Infect Dis 2004;189:2290–2293. [124] Pneumococcal conjugate vaccine for childhood immunization—WHO position paper. Wkly Epidemiol Rec 2007;82:93–104. [125] Bernstein DI, Smith VE, Sherwood JR, et al. Safety and immunogenicity of live, attenuated human rotavirus vaccine 89–12. Vaccine 1998;16:381–387. [126] Clark HF, Offit PA. Vaccines for rotavirus gastroenteritis universally needed for infants. Pediatr Ann 2004;33:536–543. [127] Clark HF, Offit PA, Ellis RW, et al. The development of multivalent bovine rotavirus (strain WC3) reassortant vaccine for infants. J Infect Dis. 1996;174(Suppl 1):S73–S80. [128] Clark HF, Offit PA, Plotkin SA, et al. The new pentavalent rotavirus vaccine composed of bovine (strain WC3)-human rotavirus reassortants. Pediatr Infect Dis J. 2006;25:577–583. [129] Patel MM, López-Collada VR, Bulhões MM, et al. Intussusception risk and health benefits of rotavirus vaccination in Mexico and Brazil. N Engl J Med 2011;364:2283–2292. [130] Yih WK, Lieu TA, Kulldorff M, et al. Intussusception Risk after Rotavirus Vaccination in U.S. Infants. N Engl J Med 2014;370: 503–512. [131] Weintraub ES, Baggs J, Duffy J, et al. Risk of intussusception after monovalent rotavirus vaccination. N Engl J Med. 2014;370:513–519. [132] Glass RI, Parashar UD. Rotavirus vaccines–balancing intussusception risks and health benefits. N Engl J Med. 2014;370:568–570. [133] Germanier R, Füer E. Isolation and characterization of Gal E mutant Ty 21a of Salmonella typhi: a candidate strain for a live, oral typhoid vaccine. J Infect Dis 1975;131:553–558. [134] Ivanoff B, Levine MM. Typhoid fever: continuing challenges from a resilient bacterial foe. Bull Inst Pasteur 1997;95:129–142. [135] Tarr PE, Kuppens L, Jones TC, et al. Considerations regarding mass vaccination against typhoid fever as an adjunct to sanitation and public health measures: potential use in an epidemic in Tajikistan. Am J Trop Med Hyg 1999;61:163–170. [136] D’Amelio R, Tagliabue A, Nencioni L, et al. Comparative analysis of immunological responses to oral (Ty21a) and parenteral (TAB) typhoid vaccines. Infect Immun 1988;56:2731–2735. [137] WHO. Cholera vaccines. WHO position paper. Weekly Epidemiological Record 2010;85: 117– 128. [138] Takahashi M. Development and characterization of a live varicella vaccine (Oka strain). Biken J. 1984;27:31–36. [139] Oxman MN, Levin MJ, Johnson GR, et al. A vaccine to prevent herpes zoster and postherpetic neuralgia in older adults” (PDF). N Engl J Med 2005;352:2271–2284. [140] Oxman MN, Levin MJ. Shingles Prevention Study Group. Vaccination against Herpes Zoster and Postherpetic Neuralgia. J Infect Dis 2008;197(Suppl 2):S228–S236. [141] Lal H, Cunningham AL, Godeaux O, et al. Efficacy of an adjuvanted herpes zoster subunit vaccine in older adults. N Engl J Med. 2015;372:2087–296. [142] Steffen R. Immunization against hepatitis A and hepatitis B infections. J Travel Med 2001;8(Suppl 1):S9–S16. [143] Hicks DJ, Fooks AR, Johnson N. Developments in rabies vaccines. Clin Exp Immunol 2012;169:199–204. [144] Macleod CM, Hodges RG, Heidelberger M, et al. Prevention of pneumococcal pneumonia by immunization with specific capsular polysaccharides. J Exp Med. 1945;82:445–465.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

28

E. D’AMELIO ET AL.

[145] Robbins JB, Parke JC Jr, Schneerson R, et al. Quantitative measurement of “natural” and immunization-induced Haemophilus influenzae type b capsular polysaccharide antibodies. Pediatr Res. 1973;7:103–110. [146] Gotschlich EC, Goldschneider I, Artenstein MS. Human immunity to the meningococcus. IV. Immunogenicity of group A and group C meningococcal polysaccharides in human volunteers. J Exp Med. 1969;129:1367–1384. [147] Wong KH, Feeley JC, Northrup RS, et al. Vi antigen from Salmonella typhosa and immunity against typhoid fever. I. Isolation and immunologic properties in animals. Infect Immun. 1974;9:348–353. [148] Le Moli S, Matricardi PM, Quinti I, et al. Clonotypic analysis of human antibodies specific for Neisseria meningitidis polysaccharides A and C in adults. Clin Exp Immunol 1991;83:460–465. [149] Lambert PH, Liu M, Siegrist CA. Can successful vaccines teach us how to induce efficient protective immune responses? Nat Med. 2005;11(4 Suppl):S54–S62. [150] Avery OT, Goebel WF. Chemical-immunological studies on conjugated carbohydrate-proteins. II. Immunological specificity of synthetic sugar-protein antigens. J Exp Med 1929; 50: 533–542. [151] Jennings H. Further approaches for optimizing polysaccharide-protein conjugate vaccines for prevention of invasive bacterial disease. J Infect Dis. 1992;165(Suppl 1):S156–S159. [152] Tai JY, Vella PP, McLean AA, et al. Haemophilus influenzae type b polysaccharide-protein conjugate vaccine. Proc Soc Exp Biol Med. 1987;184:154–161. [153] Schneerson R, Barrera O, Sutton A, et al. Preparation, characterization, and immunogenicity of Haemophilus influenzae type b polysaccharide–protein conjugates. J Exp Med 1980;152:361–376. [154] Fairley CK, Begg N, Borrow R, et al. Conjugate meningococcal serogroup A and C vaccine: reactogenicity and immunogenicity in United Kingdom infants. J Infect Dis 1996;174:1360–1363. [155] Schneerson R. Robbins JB, Park JC Jr, et al. Quantitative and qualitative analyses of serum antibodies elicited in adults by Haemophilus influenzae type b and pneumococcus type 6A capsular polysaccharide-tetanus toxoid conjugates. Infect Immun 1986;52:519–528. [156] Szu, SC, Stone AL, Robbins JD, et al. Vi capsular polysaccharide-protein conjugates for prevention of typhoid fever. J. Exp. Med 1987;166:1510–1524. [157] Pollard AJ, Perrett KP, Beverley PC. Maintaining protection against invasive bacteria with protein-polysaccharide conjugate vaccines. Nat Rev Immunol 2009;9:213–220. [158] Clutterbuck EA, Lazarus R, Yu LM, et al. Pneumococcal conjugate and plain polysaccharide vaccines have divergent effects on antigen-specific B cells. J Infect Dis 2012;205:1408–1416. [159] Clarke ET, Williams NA, Findlow J, et al. Polysaccharide-Specific memory B cells generated by conjugate vaccines in humans conform to the CD27+IgG+ isotype-switched memory B cell phenotype and require contact-dependent signals from bystander T cells activated by bacterial proteins to differentiate into plasma cells. J Immunol 2013;191:6071–6083. [160] Poolman JT, Peeters CC, van den Dobbelsteen GP. The history of pneumococcal conjugate vaccine development: dose selection. Expert Rev Vaccines 2013;12:1379–1394. [161] Moffitt KL, Malley R. Next generation pneumococcal vaccines. Curr Opin Immunol 2011;23:407–413. [162] Moffitt KL, Yadav P, Weinberger DM, et al. Broad antibody and T cell reactivity induced by a pneumococcal whole-cell vaccine. Vaccine. 2012;30:4316–4322. [163] CDC. Biological and chemical terrorism: strategic plan for preparedness and response, recommendations of the CDC Strategic Planning Workgroup 2000. MMWR Morb Mortal Wkly Rep 2000;49(RR-4):1–14. [164] Jernigan DB, Raghunathan PL, Bell BP, et al. National Anthrax Epidemiologic Investigation Team. Investigation of bioterrorism-related anthrax, United States, 2001: epidemiologic findings. Emerg Infect Dis 2002;8:1019–1028. [165] Reed DS, Smith LP, Cole KS, et al. Live attenuated mutants of Francisella tularensis protect rabbits against aerosol challenge with a virulent type A strain. Infect Immun. 2014;82:2098–2105. [166] Rueckert C, Guzmán CA. Vaccines: from empirical development to rational design. PLoS Pathog 2012;8:e1003001. [167] Plotkin SA. Six revolutions in vaccinology. Pediatr Infect Dis J 2005;24:1–9. [168] Plotkin SA. Minireview. Vaccines: the fourth century. 2009;16:1709–1719. [169] Neumann G, Watanabe T, Ito H, et al. Generation of influenza A viruses entirely from cloned cDNAs. Proc Natl Acad Sci USA 1999;96:9345–9350.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

29

[170] Mora M, Veggi D, Santini L, et al. Reverse vaccinology. Drug Discov Today 2003;8:459–464. [171] Moriel DG, Scarselli M, Serino L, et al. Genome-based vaccine development: a short cut for the future. Hum Vaccin. 2008;4:184–188. [172] Kelly DF, Rappuoli R. Reverse vaccinology and vaccines for serogroup B Neisseria meningitidis. Adv Exp Med Biol 2005;568:217–223. [173] Giuliani MM, Adu-Bobie J, Comanducci M, et al. A universal vaccine for serogroup B meningococcus. Proc Natl Acad Sci USA 2006;103:10834–10839. [174] Flingai S, Czerwonko M, Goodman J, et al. Synthetic DNA vaccines: improved vaccine potency by electroporation and co-delivered genetic adjuvants. Front Immunol 2013;4:354. [175] Pulendran B, Li S, Nakaya HI. Systems vaccinology. Immunity 2010;33:516–529. [176] Nakaya HI, Li S, Pulendran B. Systems vaccinology: learning to compute the behavior of vaccine induced immunity. Wiley Interdiscip Rev Syst Biol Med. 2012;4:193–205. [177] Lessons from vaccine history. Nat Med 2012;18:1717. [178] Halstead SB. Pathogenesis of dengue: challenges to molecular biology. Science 1988;239:476– 481. [179] Villar L, Dayan GH, Arredondo-García JL et al. Efficacy of a tetravalent dengue vaccine in children in Latin America. N Engl J Med. 2015;372:113–123. [180] Bhatt S, Gething PW, Brady OJ, et al. The global distribution and burden of dengue. Nature 2013;496:504–507. [181] WHO. Hepatitis C. Fact sheet N°164, updated April 2014. Available at www.who.int/mediacentre/ factsheets/fs164/en/ [182] Cox AL, Thomas DL. Hepatitis C virus vaccines among people who inject drugs. Clin Infect Dis 2013;57(S2):S46–S50. [183] WHO, Fact sheet N°280, updated June 2014. Available at www.who.int/mediacentre/factsheets/ fs280/en/ [184] Zhang J, Zhang XF, Huang SJ, et al. Long-term efficacy of a hepatitis E vaccine. N Engl J Med. 2015;372:914–922. [185] Flynn NM, Forthal DN, Harro CD, et al. Placebo-controlled phase 3 trial of a recombinant glycoprotein 120 vaccine to prevent HIV-1 infection. J Infect Dis. 2005;191:654–665. [186] Pitisuttithum P, Gilbert P, Gurwith M, et al. Randomized, double-blind, placebo-controlled efficacy trial of a bivalent recombinant glycoprotein 120 HIV-1 vaccine among injection drug users in Bangkok, Thailand. J Infect Dis. 2006;194:1661–1671. [187] Buchbinder SP, Mehrotra DV, Duerr A, et al. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet 2008;372:1881–1893. [188] Gray G, Buchbinder S, Duerr A. Overview of STEP and Phambili trial results: two phase IIb test-of-concept studies investigating the efficacy of MRK adenovirus type 5 gag/pol/nef subtype B HIV vaccine. Curr Opin HIV AIDS 2010;5:357–361. [189] Gray GE, Allen M, Moodie Z, et al. Safety and efficacy of the HVTN 503/Phambili study of a clade-B-based HIV-1 vaccine in South Africa: a double-blind, randomised, placebo-controlled test-of-concept phase 2b study. Lancet Infect Dis. 2011;11:507–515. [190] Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med. 2009;361:2209–2220. [191] Haynes BF, Gilbert PB, McElrath MJ, et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med. 2012;366:1275–1286. [192] Walker LM, Huber M, Doores KJ, et al. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 2011;477:466–470. [193] Burton DR, Poignard P, Stanfield RL, et al. Broadly neutralizing antibodies present new prospects to counter highly antigenically diverse viruses. Science 2012;337:183–186. [194] Corti D, Lanzavecchia A. Broadly neutralizing antiviral antibodies. Annu Rev Immunol. 2013;31:705–742. [195] Nabel GJ. Designing Tomorrow’s vaccines. N Engl J Med 2013;368:551–560. [196] Wu X, Zhou T, Zhu J, et al. Focused evolution of HIV-1 neutralizing antibodies revealed by structures and deep sequencing. Science 2011;333:1593–1602. [197] Dimitrov DS. Therapeutic antibodies, vaccines and antibodyomes. mAbs 2010;2:347–356.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

30

E. D’AMELIO ET AL.

[198] Kim HW, Canchola JG, Brandt CD, et al. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am. J. Epidemiol 1969;89:422–434. [199] Dudas RA, Karron RA. Respiratory Syncytial Virus Vaccines. Clin Microbiol Rev 1998;11:430– 439. [200] Gomez RS, Guisle-Marsollier I, Bohmwald K, et al. Respiratory Syncytial Virus: pathology, therapeutic drugs and prophylaxis. Immunol Lett. 2014;162(1 Pt A):237–247. [201] Kabanova A, Perez L, Lilleri D, et al. Antibody-driven design of a human cytomegalovirus gHgLpUL128L subunit vaccine that selectively elicits potent neutralizing antibodies. Proc Natl Acad Sci U S A. 2014;111:17965–17970. [202] Worgall S. 40 years on: have we finally got a vaccine for Pseudomonas aeruginosa? Future Microbiol. 2012;7:1333–1335. [203] Bagnoli F, Fontana MR, Soldaini E, et al. Vaccine composition formulated with a novel TLR7dependent adjuvant induces high and broad protection against Staphylococcus aureus. Proc Natl Acad Sci U S A.2015;112:3680–3685. [204] Murray CJ, Ortblad KF, Guinovart C, et al. Global, regional, and national incidence and mortality for HIV, tuberculosis, and malaria during 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2014;384:1005–1070. [205] Agnandji ST, Lell B, Soulanoudjingar SS, et al. First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. N Engl J Med. 2011;365:1863–1875. [206] Henao-Restrepo AM, Longini IM, Egger M, et al. Efficacy and effectiveness of an rVSV-vectored vaccine expressing Ebola surface glycoprotein: interim results from the Guinea ring vaccination cluster-randomised trial. The Lancet 2015;[Epub ahead of print]:pii:S0140-6736(15)61117-5. [207] WHO. Ebola vaccines, therapies and diagnostics. Questions and answers.2015. Last update 27 May 2015. Available at www.who.int/medicines/emp_ebola_q_as/en/ [208] Rebeaud F, Bachmann MF. Immunization strategies for Clostridium difficile infections. Expert Rev Vaccines. 2012;11:469–479. [209] Spencer J, Leuzzi R, Buckley A, et al. Vaccination against Clostridium difficile using toxin fragments: observations and analysis in animal models. Gut Microbes. 2014;5:225–232. [210] Ramon G. Procédés pour accroitre la production des Antitoxines. Ann Inst Pasteur 1926;40: 1–10. [211] Kuroda E, Coban C, Ishii KJ. Particulate adjuvant and innate immunity: past achievements, present findings, and future prospects. Int Rev Immunol 2013;32:209–220. [212] Hogenesch H. Mechanism of immunopotentiation and safety of aluminum adjuvants. Front Immunol 2013;3:406. [213] Watson DS, Endsley AN, Huang L. Design considerations for liposomal vaccines: influence of formulation parameters on antibody and cell-mediated immune responses to liposome associated antigens. Vaccine. 2012;30:2256–2272. [214] Tagliabue A, Rappuoli R. Vaccine adjuvants. The dream become real. Human Vaccines 2008;4:347–349. [215] Khurana S, Chearwae W, Castellino F, et al. Vaccines with MF59 adjuvant expand the antibody repertoire to target protective sites of pandemic avian H5N1 influenza virus. Sci Transl Med 2010;2:15ra5. [216] Khurana S, Verma N, Yewdell JW, et al. MF59 adjuvant enhances diversity and affinity of antibody-mediated immune response to pandemic influenza vaccines. Sci Transl Med 2011;3:85ra48. [217] Tsai TF. MF59 adjuvanted seasonal and pandemic influenza vaccines. Yakuga Zasshi 2011;131:1733–1741. [218] Milanetti F, Germano V, Nisini R, et al. Safety and immunogenicity of co-administered MF59adjuvanted 2009 pandemic and plain 2009–10 seasonal influenza vaccines in rheumatoid arthritis patients on biologicals. Clin Exp Immunol 2014;177:287–294. [219] Miller E, Andrews N, Stellitano L, et al. Risk of narcolepsy in children and young people receiving AS03 adjuvanted pandemic A/H1N1 2009 influenza vaccine: retrospective analysis. BMJ 2013;346:f794. [220] Tsai T, Del Giudice G, Crucitti A, et al. Is the adjuvant solely to blame? BMJ. 2013;346:f2375.

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

INTERNATIONAL REVIEWS OF IMMUNOLOGY

31

[221] Masoudi S, Ploen D, Kunz K, et al. The adjuvant component α-tocopherol triggers via modulation of Nrf2 the expression and turnover of hypocretin in vitro and its implication to the development of narcolepsy. Vaccine. 2014;32:2980–2988. [222] Plotkin SA, Orenstein WA, Offit PA. Vaccines. 5th Edition. Philadelphia: Saunders, Elsevier; 2008. [223] Calabro S, Tortoli M, Baudner BC, et al. Vaccine adjuvants alum and MF59 induce rapid recruitment of neutrophils and monocytes that participate in antigen transport to draining lymph nodes. Vaccine 2011;29:1812–1823. [224] Didierlaurent AM, Morel S, Lockman L, et al. AS04, an aluminum salt-and TLR4 agonist- based adjuvant system, induces a transient localized innate immune response leading to enhanced adaptive immunity. J Immunol 2009;183:6186–6197. [225] Glück R, Mischler R, Brantschen S, et al. Immunopotentiating reconstituted influenza virus virosome vaccine delivery system for immunization against hepatitis A. J Clin Invest 1992;90:2491– 2495. [226] Morel S, Didierlaurent A, Bourguignon P, et al. Adjuvant system AS03 containing α-tocopherol modulates innate immune response and leads to improved adaptive immunity. Vaccine 2011;29:2461–2473. [227] Moser C, Muller M, Kaeser MD, et al. Influenza virosomes as vaccine adjuvant and carrier system. Expert Rev Vaccines 2013;12:779–791. [228] Awate S, Babiuk LA, Mutwiri G. Mechanisms of action of adjuvants. Front Immunol. 2013;4:114. [229] Lee S, Nguyen MT. Recent advances of vaccine adjuvants for infectious diseases. Immune Netw. 2015;15:51–57. [230] Nakaya HI, Wrammert J, Lee EK, et al. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol. 2011;12:786–795. [231] MMWR. Global routine vaccination coverage. 2013.2014;63:1055–1058. [232] Steffen R, Connor BA. Vaccines in travel health: from risk assessment to priorities. J Travel Med 2005;12:26–35. [233] D’Amelio R, Heymann DL. Can the military contribute to global surveillance and control of infectious diseases? Emerg Infect Dis 1998;4:704–705. [234] Grabenstein JD, Pittman PR, Greenwood JT, et al. Immunization to protect the US Armed Forces: heritage, current practice, and prospects. Epidemiol Rev 2006;28:3–26. [235] Bozzo P, Narducci A, Einarson A. Vaccination during pregnancy. Can Fam Physician 2011; 57:555–557. [236] Sydnor E, Perl TM. Healthcare providers as sources of vaccine-preventable diseases. Vaccine. 2014;32:4814–4822. [237] Fortunato F, Tafuri S, Cozza V, et al. Low vaccination coverage among Italian healthcare workers in 2013. Hum Vaccin Immunother. 2015;11:133–139. [238] Thompson CN, Kama M, Acharya S, et al. Typhoid fever in Fiji: a reversible plague? Trop Med Int Health. 2014;19:1284–1292. [239] Yu MI, Lee SD, Lu RH, et al. Need for vaccination of susceptible food handlers against hepatitis A in Taiwan. Zhonghua Yi Xue Za Zhi (Taipei). 2000;63:798–803. [240] Germano V, Cattaruzza MS, Osborn J, et al. Infection risk in Rheumatoid Arthritis and Spondyloarthropathy patients under treatment with DMARDs, Corticosteroids and TNF-α antagonists. J Transl Med 2014;12:77. [241] Salemi S, D’Amelio R. Are anti-infectious vaccination safe and effective in patients with autoimmunity? Int Rev Immunol 2010;29:270–314. [242] Hmamouchi I, Winthrop K, Launay O, et al. Low rate of influenza and pneumococcal vaccine coverage in rheumatoid arthritis: Data from the international COMORA cohort. Vaccine. 2015;33:1446–1452. [243] van Assen S, Agmon-Levin N, Elkayam O, et al. EULAR recommendations for vaccination in adult patients with autoimmune inflammatory rheumatic diseases. Ann Rheum Dis. 2011;70:414–422. [244] Dougados M. Low rate of influenza and pneumococcal vaccine coverage in rheumatoid arthritis: data from the international COMORA cohort. Vaccine. 2015;33:1446–1452. [245] Biondo MI, Germano V, Pietrosanti M, et al. Lack of hepatitis B virus reactivation after antitumour necrosis factor α agents therapy in antibody to hepatitis B core antigen positive/ hepatitis

32

[246] [247] [248] [249] [250]

Downloaded by [University of California Santa Barbara] at 23:37 26 November 2015

[251] [252] [253] [254] [255] [256] [257] [258] [259] [260]

E. D’AMELIO ET AL.

B surface antigen negative subjects with chronic inflammatory arthropathies. Eur J Intern Med. 2014;25:482–484. Ferreira I, Isenberg D. Vaccines and biologics. Ann Rheum Dis. 2014;73:1446–1454. Weinberger B, Grubeck-Loebenstein B. Vaccines for the elderly. Clin Microbiol Infect 2012;18(Suppl 5):100–108. Centers for Disease Control and Prevention. Biological and chemical terrorism: strategic plan for preparedness and response, recommendations of the CDC Strategic Planning Workgroup 2000. MMWR Morb Mortal Wkly Rep 2000;49(RR-4):1–14. Ehreth J. The global value of vaccination. Vaccine 2003;21:596–600. Zenteno-Cuevas R. Update on the Development of TB vaccines. Curr Pharm Biotechnol. 2013; 14:940–946. Frey SE, Winokur PL, Salata RA, et al. Safety and immunogenicity of IMVAMUNE® smallpox vaccine using different strategies for a post event scenario. Vaccine 2013; 31:3025–3033. Oyston PC, Williamson ED. Prophylaxis and therapy of plague. Expert Rev Anti Infect Ther 2013;11:817–829. Esparza J. A brief history of the global effort to develop a preventive HIV vaccine. Vaccine 2013;31:3502–3518. Yauch LE, Shresta S. Dengue virus vaccine development. Adv Virus Res 2014;88:315–372. Yang K, Varga SM. Mucosal vaccines against respiratory syncytial virus. Curr Opin Virol 2014;6:78–84. Wang D, Fu TM. Progress on human cytomegalovirus vaccines for prevention of congenital infection and disease. Curr Opin Virol 2014;6C:13–23. Shedlock DJ, Aviles J, Talbott KT, et al. Induction of broad cytotoxic T cells by protective DNA vaccination against Marburg and Ebola. Mol Ther. 2013;21:1432–1444. Low N, Bavdekar A, Jeyaseelan L, et al. A randomized, controlled trial of an aerosolized vaccine against measles. N Engl J Med 2015;372:1519–1529. Offit P. Back to the future. Expert Rev Vaccines 2004;3:107–108. Yang YT, Silverman RD. Social distancing and the unvaccinated. N Engl J Med 2015;372:1481– 1483.

Anti-Infectious Human Vaccination in Historical Perspective.

A brief history of vaccination is presented since the Jenner's observation, through the first golden age of vaccinology (from Pasteur's era to 1938), ...
566B Sizes 2 Downloads 20 Views