JCDR - Register at Journal of Clinical and Diagnostic Research
Journal of Clinical and Diagnostic Research, ISSN - 0973 - 709X
Microbiology Section DOI : 10.7860/JCDR/2019/41441.13071
Year : 2019 | Month : Aug | Volume : 13 | Issue : 08 Full Version Page : DE01 - DE07

Vaccine Development Against Salmonella Typhi: A Search is Still On

Shailendra Singh1, Chandra Bhan Pratap2, Gopal Nath3

1 PhD Scholar, Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
2 Postdoctoral Fellow, Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
3 Professor, Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India.


NAME, ADDRESS, E-MAIL ID OF THE CORRESPONDING AUTHOR: Dr. Gopal Nath, Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
E-mail: gopalnath@gmail.com
Abstract

Salmonella serovar typhi still remains a serious problem in South Asia, South-East Asia, and sub-Saharan Africa. The emergence of multidrug resistance, lack of proper diagnosis and chronic typhoid carrier are the main causes of such a high level of morbidity and mortality. Presently, three vaccines have been licensed for typhoid infection but none of them is optimum for complete eradication for want of safety and long lasting immunity. Several subunit and attenuated candidate vaccines are under trial. Recently, in India Typbar TCV conjugate vaccine has been licensed. However, this vaccine being subunit may not induce appropriate immune response and also it cannot be given to infants. We need a multivalent attenuated vaccine which can induce both cellular as well as humoral immunity against different pathogenic serovar of Salmonella. Live attenuated vaccine could be an attractive choice taking care of all the above points. In this review, we focus on the pathogenesis of Salmonella serovar typhi, role of humoral as well as Cell-Mediated Immune (CMI) response, different licensed vaccines with their pros and cons, and also the targets which are already been put on clinical trials. We have discussed the attenuation of the candidates by modification of certain structural and functional genes especially looking for induction of CMI.

Keywords

Introduction

Enteric fever is an acute generalised infection caused by human restricted Salmonella enterica subspecies enterica serovar Typhi, Paratyphi A and B. Majority (90%) of enteric fever cases are caused by Typhi serotypes. Enteric fever is a very important public health problem in countries with poor sanitation and inadequate hygiene. The recent global burden of typhoid fever is estimated to be ranging between 11 and 21 million. This burden is associated with 128000 to 161000 deaths annually [1]. The most affected age group for enteric fever is less than 14 years [2]. The most disturbing issue is that about 27% of the infections occur in the age group <4 years of which 10% are in infants <1 year of age. Typhoid is a major threat for the Indian subcontinent due to its high prevalence, severity, development of Multidrug Resistant (MDR) and persistence of carrier state in our population. Approximately, 2-5% of cases as well as subclinical infections develop carrier state irrespective of preexisting gall bladder disease [3]. The carrier state has been implicated in biliary tract cancer [4]. Chronic carriers are the reservoir of infection and are responsible for endemicity of the fever. Following ingestion, the organisms apart from resisting the low pH of the stomach, they get signals for induce multiplication before reaching the ileum [5]. In the small intestine, the bacteria multiply in the submucosa and Peyer’s patches [6]. During systemic infection, bacteria reach the phagocytes of spleen, liver and bone marrow. Later, the infection manifests as bacteraemia. Death due to typhoid fever occurs usually due to perforation of the intestine or haemorrhage.

Chloramphenicol was introduced in 1948 and their resistance started to develop within two years of introduction of the drug and until 1972 chloramphenicol resistant S. typhi became a major problem [7]. Outbreaks of chloramphenicol resistant Salmonella occurred in Mexico, India, Vietnam, Thailand, Korea and Peru. The case fatality rate without antibiotic therapy was 10-20%. However, with antibiotics, it has come down to 1-4%. This mortality rate is usually due to delay in start of therapy. After the suggestion of Threfall JE et al., that S. typhi with decreased sensitivity to ciprofloxacin is endemic in several Asian countries [8], there has been several reports indicating that quinolones have almost lost their ground as a magic drug [9]. A gradual increase in mean Minimum Inhibitory Concentration (MIC) of ceftriaxone has already been reported by our group recently [9]. Emergence of resistance against the two second line drugs; ciprofloxacin and ceftriaxone has become a serious problem in the recent time. Recently, azithromycin is also showing the resistance as 20% of the recent isolates of S. typhi was showed unresponsive [10]. Then what can be done? Bacteriophage therapy (still under preliminary phase of development) or newer antibiotic molecules (which takes long time to reach clinical application) may be an alternative to the existing antibiotics. Immunotherapy is not practicable at the community level. So logically, a perfect or near perfect vaccine may be able to eradicate this human restricted bacteria globally.

It is important to understand the host immune response against the bacteria before addressing the issue of vaccine development. The subclinical, clinical and chronic carrier states basically depend on the balance between the virulence of the invader and the immune response of the host. The immunity against the bacterium is divided in two parts:

Innate Immunity against Salmonella

Components of innate immune system viz., complement and opsonising serum antibodies facilitate the uptake of the bacteria by phagocytes (macrophage, neutrophils and dendritic cells) [11] and epithelial cells and help to identifying specific Pathogen-Associated Molecular- Patterns (PAMPs) of the pathogen. In Salmonellosis, TLR 4, TLR5 and TLR9 signaling systems are activated by bacterial DNA, flagella and Lipopolysaccharide (LPS). Salmonellaenterica species is adapted to mammals including humans and has a complex cross-talk system resulting ultimately in induction of host immune response. The bacteria are seeded into different organs of the body viz., liver, gallbladder, spleen, bone marrow etc., during sustained bacteraemia. A cycle of infection continues and bacteria either reinvade epithelial cells of the intestinal wall or shed in the faeces. However, after a certain period, the symptoms of Salmonellosis resolve. A small population of hosts may become chronic carriers [3,12]. Proinflammatory cytokine (IL-1β, IL-6, IFN-γ and TNF-α) are recruited to promote systemic inflammation [13,14]. IFN-γ is a known macrophage activating factor. It plays an important role in persistence of infection. Macrophage activity and different type of cytokine secretion is primarily involved in the induction of innate as well as adaptive immune responses. Further, equilibrium between pro-inflammatory and anti-inflammatory cytokines is maintained which controls the infection along with ensuring minimal damage to the host. Bone marrow derived macrophages and primary cell line have shown that Salmonella promote chemokine and cytokine synthesis in macrophages, dendritic and epithelial cells [15]. In typhoid fever cases, an aggressive pro-inflammatory response against Salmonella is not a common occurrence. However, in patients suffering from typhoid fever, distinct peripheral blood metabolite profile has been elucidated by microarray and transcription profiling techniques [14, 16]. It is interesting to note that if the hosts are incapable of mounting an appropriate response, they are more prone to relapse, re-infection and occasionally becoming chronic carriers [16]. There is scarcity of data of host immune defence in human beings against Salmonella infection. In mouse tissue, early invasion of Salmonella is controlled by the innate resistance by autosomal gene Nramp1 (Natural resistance associated macrophage protein 1) which functions as a divalent metal ion pump. This gene is expressed mainly in macrophages and cells of granulocyte lineage [17]. It has been stated that the main host defence against Salmonella is exhibited through neutrophils followed by mononuclear cells. Clearance of bacteria from tissue requires activation of CD4+ and TCR-αβ T-cells. This activation depends on CD28 and is controlled by MHC class II genes [18,19]. Dendritic cells and B-cells are involved in initiation and development of T-cell immunity to Salmonella [20]. Salmonella is a facultative intracellular organism; therefore, apart from antibody, cell mediated immunity through B and T-cell cooperation, Th1 type immunological memory and CD8+ T-cells may essentially be required.

Humoral Immune Response

There are studies showing antibody response to LPS and protein determinants in mice [21], rabbits [22] and humans [23] following exposure to live or killed Salmonella. Serum and mucosal IgA has usually been found to be induced after vaccination and also by some of the killed preparations [21]. However, it has been speculated that antibodies alone may not provide immunity against re-infection and recurrence.

Cell Mediated Immune Response

Delayed Type of Hypersensitivity (DTH) response not only against proteinaceous antigens, porins but also to LPS antigen determinants and Vi surface polysaccharide has been observed in mice [24] after immunisation with live vaccine. However, DTH could not be detected after immunisation with killed vaccine. Intracellular bacteria have a challenge to maintain a balance of weakened host to prevent clearance along with the healthy enough to establish a suitable niche for their existence and intracellular replication. This is the basic essential requirement for human restricted pathogen like Salmonella typhi serotypes. Such pathogens modulate host intracellular signaling in their own favour to consume resources and neutralise the host defence mechanisms. Moreover, different bacteria have developed individualised strategies to target the same host cell. The killing of intracellular bacteria is by phagocytosis in phagosomes which is followed by fusion with the lysosome to give phagolysosomes. The pH of phagolysosome is 3.5-4.0 which is toxic to bacteria. The killing is further enhanced by acid proteases. The surviving facultative Salmonella bacteria inside the Salmonella-Containing Vacuoles (SCV) express certain genes (spiC, sseB) which inhibit the fusion of phagosome to lysosome.

Past and Currently Available Vaccines

Vaccination is a powerful technique for mitigating Salmonella related problems. In the past, two methods were used for vaccine development: attenuation (heat, oxygenation, chemical agents or old culture) and inactivation by killing the bacteria. Subunit vaccines have also been developed and used against Salmonella with variable success. Killed vaccine failed due to some reactogenicity and poor immunity. Attenuated vaccine has also been tried with variable success. The two vaccines are available in clinical practice against typhoid. However, they are not satisfactory in prevention of the disease [25]. These two vaccines are, live attenuated Ty21a and Vi capsular polysaccharide derived [26]. Recently, a new vaccine has been approved by WHO (World Health Organisation) which recommended for use in areas with a high burden of typhoid and higher antibiotic resistance [1].

Whole Cell Inactivated Vaccine

Initially, whole cell inactivated vaccine was used for prevention of typhoid fever which provided 73% protection for three years. Unfortunately, this vaccine was reactogenic. Therefore, this vaccine is no longer recommended. This was a heat killed phenol preserved or acetone killed lyophilized vaccine derived from whole bacteria. Heat killed vaccine is no better than acetone killed vaccine because heat may destroy protein conformation as well as the Vi polysaccharide which are immunogenic otherwise [27]. However, inactivated whole cell vaccines can cause inflammation, pain fever and other side effects which are not suitable for mass vaccination [28]. Killed vaccine can induce sufficient humoral immunity but they are unable to induce Th1 or in some case even, Th2 mediated immune response [21,29]. In case of humans, killed vaccine gave better protection when given parenterally than orally.

Vi-Vaccine and Other Subunit Vaccines

Vi antigen on Salmonella serovar Typhi gives protection from complement-mediated bacterial lysis and also inhibits alternative complement activation pathway. Vi polysaccharide is purified from the Ty21 strain of serovar Typhi. Vi vaccine is an unconjugated purified capsular polysaccharide and it has provided 77% protection for 20 months and it is prohibited for children below two years of age. A single dose of Vi vaccine is sufficient to induce a high titre of anti-Vi antibody. Felix A et al., (1935) for the first time showed that Vi polysaccharide of Salmonella typhi induces humoral immune response against typhoid [30]. It is a linear homopolymer of galacturonic acid isolated from Salmonella typhi cetavlon detergent treatment. This vaccine was first licensed in USA in 1994 and is available globally. Vi antigen does not induce a T-cell response because it is a polysaccharide and therefore no affinity maturation and memory cells development. Further Vi vaccine does not provide protection against Vi negative S. typhi and Paratyphi serotypes of Salmonellaenterica and it needs re-vaccination two years after the initial dose. The Vi vaccine gives 55% to 75 % protection in typhoid endemic areas. Their protection rate varied in different age groups and countries such as 75% protection rate in five to seven-year-old for 20 months in Nepal, 64% in 5-16-year-old in South Africa for 21 months whereas it came down to 55% after 36 months, while in China 69% protection in 6-9 year old children [31,32]. In children below 2 years of age even after booster, it does not induce immunity. Another drawback is that all strains of Salmonella typhi do not express Vi polysaccharide or hide their Vi polysaccharide on their surface and so it is unable to prevent typhoid [33]. Immunogenicity was found to increase when it was conjugated with a carrier protein such as tetanus toxoid, diphtheria toxin, porin or recombinant exoprotein of Pseudomonas aeruginosa [34].

Live Attenuated Ty21a Vaccine

Due to the presence of adverse effects of heat killed vaccine, a live attenuated vaccine was required which could induce prolonged immunity against typhoid. Ty21a, the first oral live attenuated vaccine against typhoid was developed in a wild type strain by chemical mutagenesis [35]. In case of Ty21a, two genes galE and Vi were mutated randomly yielding highly attenuated strain of Salmonella typhi. The galE gene encodes UDP galactose-4-epimerase and Vi gene encodes surface polysaccharide. Once this gene is mutated, the mutant produces rough LPS in place of smooth LPS. When galactose is exogenously supplied the mutant was able to make smooth LPS till it was the available. This smooth LPS can protect the bacterium from phagocytosis. This vaccine gives protection via induction of anti-O and anti-H antibody (mucosal and serum) and also up to some extent of cellular immunity. Cellular immunity is induced in the form of T-cell activation and cytokine secretion. Currently this vaccine has been licensed in 56 countries of Africa, USA, Europe and Asia [36]. However, the immunity induced by this vaccine diminishes with increasing duration after vaccination. The protection rate differs from region to region viz., in Egypt it has given 96% protection for three years whereas in Chile, only 67% protection for three years. Later the protection rate decreased to 62 % for 7 years. The protection rate also varies in different age groups i.e. 59% in 5-9 years, 67% in 10-14 years and 85% in more than 15 years of age. Interestingly this chemically mutated vaccine also gives cross-protection against paratyphoid fever (Paratyphi B) causing serotypes but their protection level was only 42-56 %. This vaccine is also able to develop herd immunity in endemic settings.

Typbar-TCV (Typhoid Conjugate Vaccine)

It is developed by conjugation of tetanus toxoid protein with purified Vi polysaccharide (Vi-TT). It was licensed for the first time in India in 2013. It is given to children more than 6 months of age. Typbar TCV induces a high titre of anti-Vi antibody as well as higher avidity in comparison to Vi–unconjugated vaccine. Protection persisted in 84% infants (6-23-month-old) for 5 years. The efficacy in human volunteers after one month has been reported to be 87.1% and sero-efficacy 85% [37]. [Table/Fig-1] shows the status of current typhoid vaccines [25,26,38-43].

Status of current typhoid vaccines [25,26,38-43].

Vaccine characteristicsTy21a vaccineVi polysaccharide vaccine (ViPS)Typhoid conjugate vaccine (TCV)
TypeLive, attenuated (Chemically induced mutagenesis affecting several genes)Subunit (Purified Vi polysaccharide from Ty2 S. typhi strain)Conjugated subunit vaccine (purified Vi polysaccharide conjugate with tetanus toxoid, recombinant Exo Protein A and recombinant Diphtheria Toxin) [41,43]
WHO recommended(2008) endemic and epidemic settings(2008) endemic and epidemic settings(2013) High typhoid burden or high antimicrobial resistance
Storage and preservative2-8° C to for 3 years2-8° C for 3 yearsPhenol (1.250 mg) per dose2-8° C2-Phenoxyethanol (5 mg)
Type of immune inductionIt induces serum and mucosal immunity against both O (somatic) and H (flagellar) antigensSingle dose induces higher anti-Vi antibody titre in serum.Single dose elicited higher anti-Vi antibody titre in serum
Licensed1983 in Europe,1989 in USA1994 in USA2013 in India
Age of vaccinationEnteric coated capsules >6 years as per manufacturer≥2 years≥6 months
ScheduleEnteric coated capsules: 3-4 doses on alternate daysSingle doseSingle dose
AdministrationOral0.5ml injection (Intramuscularly or subcutaneously0.5 ml injection (Intramuscularly)
Efficacy33-77% [25].55-72% [25].85-87.1% [41,43].
Duration of protection5-7 years2-3 yearsUp to 5 years
Cross protection against NTSPartial protection against Paratyphi B (49%) [39].No evidence of cross protectionNot studied
Co-administrationIt is given with Live vaccine of polio, cholera, yellow fever and MMR- It is given as routine vaccine in children.- Hepatitis A and yellow fever in traveler.Vaccine of Mumps, Rubella and Measles show no interference with TCV [43].
Herd immunityHerd immunity developsNot applicableNot applicable
Persistence of immunityProtection from 7th day to 7-yearsProtection from 7th day to 3-yearsNot clear
Immunity in immuno-compromisedNot recommended specially CMI deficientSafeNo data available
Immunity in pregnancyNo data from pregnant women regarding these three vaccines
DisadvantageNot licensed for infants, Require 3-4 doses, Lack of anti Vi antibodyNot licensed for infants, lacks memory response, lacks affinity maturation, Only protects against S. typhi, revaccination after every 3 year.Protection only against S. typhi
Adverse effectsTransient fever and gastrointestinal problems.No serious adverse effects known.Fever, pain, swelling reported in 10% of cases.
ReferencesFraser A, et al., 2007 [25]; Poolman J, et al., 2011 [38]Khan MI, et al 2010 [26]; Kantele A, et al., 2012 [39]; Sur D, et al., 2009 [40]Mai NL, et al., 2003 [41]; Thiem VD, et al.2011 [42]; Szu SC, et al., 2013 [43]

Newer Vaccine Development

What should be the criteria for ideal Salmonella vaccine?

For an ideal vaccine against enteric fever the recommended criterion are:

Vaccine strain should not be resistant to any antibiotics.

It should be highly protective against intestinal as well as systemic infection of Salmonella typhi and should also protect against other serotypes of Salmonella.

It should be attenuated to protect humans.

Live attenuated vaccine should not cause any harm or inhibit the growth of the vaccine.

Vaccine strain should not interfere with Salmonella detection methods.

Vaccine should have a marker to differentiate it from wild type of strains.

It should easily be administered.

Live attenuated vaccine should inhibit the colonisation of other Salmonella serovars in the intestine.

In the light of above mentioned parameters, candidate vaccines are being searched in the following three directions:

Subunit vaccine (protein derived vaccine)

Carbohydrate conjugated vaccine

Live attenuated vaccine

i. Subunit vaccine (protein derived vaccine)

Subunit vaccine consists of purified recombinant protein from the targeted bacteria and, are an alternative to carbohydrate conjugated and live attenuated vaccine. The subunit vaccines are designed with bioinformatics tools that should be targeted at selection of proteins that can induce both humoral and CMI and also provide cross protection against other enteric fever causing serotypes. For designing better subunit vaccines, we must know the Salmonella epitopes on B and T-cells to enhance the potency of the protein derived subunit vaccines [44]. Protein based subunit vaccines that are under development are given below:

Flagellin and Porins, OmpC, OmpF and OmpD

OmpR-envZ is a two-component system of Salmonella in which OmpR has a response regulator of OmpC, OmpF and Vi antigen expression while EnvZ is membrane sensor protein which senses different environmental condition such as osmolarity in the surrounding [45]. The OmpC and OmpF proteins can induce a long lasting humoral response against Salmonella in a mouse model and in humans. It has been found safe and immunogenic in phase-1 clinical trials. Since these are membrane proteins, their conformation is very important for vaccine development. Protein based vaccines are non-infectious while attenuated vaccines have the possibility of reverting back to virulent form.

ii) Carbohydrate Conjugated Vaccine

Conjugated carbohydrate vaccine has more beneficial effect over pure Vi capsular polysaccharide vaccine. Polysaccharide antigen attached with carrier protein by covalent linkage can activate both arms of immune system i.e., T-cell mediated CMI and B-cell mediated antibody response. If we conjugate LPS O- antigen O: 1,2,12 with carrier protein it gives protection against S. Paratyphi-A, S. Typhimurium and S. Enteritidis O: 1,9,12 sharing O-12 antigen [46]. Generally none of the Salmonella proteins are used as a carrier. The Tetanus toxoid (TT), Diphtheria toxoid (DT) and recombinant diphtheria toxin (CRM197) are nontoxic and commonly used. Sometime rEPA (recombinant exoprotein A from Pseudomonas sp.) is also used as carrier protein for glycoconjugate vaccine. This carbohydrate conjugate carrier protein converts the antigen processing from T-independent to T-dependent antigen which may induce both humoral immunity as well as CMI. TT covalently attached with Vi antigen and rEPA carrier protein attached with Vi antigen has already been used in China and India.

If the Salmonella protein is used as carrier protein instead of exogenous protein which are shared amongst different serovars, it may provide immunity against many of them [44]. It has been reported that Salmonella O:4 conjugate with porin and O:4,5,9 conjugate with flagellin are more effective in comparison to lone porin or O:4 DT or flagellin [44,47]. Vi-CRM 197 glycoconjugate vaccine developed by Novartis Vaccine Institute for Global Health and NVGH has been tested in phase-2 clinical trials in children, infants and adults in India, Pakistan and the Philippines and phase-1 and phase-2 in adults in Europe [48]. Drawbacks of this glycoconjugate vaccine (Vi-DT or Vi-TT) are that they do not protect from enteric fever caused by Salmonella Paratyphi A or other strains because they lack the Vi antigen on their surface. On the contrary, the O:2 TT glycoconjugate vaccine is safe as well as immunogenic and target S. Paratyphi A [49]. Other O:2 DT and O:2 CRM197 glycoconjugate vaccine are under preclinical trial at the IVI (International Vaccine Institute, South Korea) and NVGH [50,51]. GelSite-OAcTM is a new derivative vaccine of Vi. In this vaccine O-acetylated heavy molecular weight polygalactouronic acid residue is attached to induce better immunity as well as memory response. Their boosting effect is effective for children under 2 years of age [52].

iii) Live attenuated Vaccine under Development

When molecular biology was not evolved to the present level, all vaccines were developed through chemical mutagenesis. However, with the development of molecular biology, specific genes can be mutated by homologous recombination as well as one step gene inactivation (site-directed mutagenesis) and selected by antibiotic resistance and checked for their live attenuated status and potential to prevent the disease. Later, the antibiotic resistance gene may be deleted from the mutant strain [53]. Live attenuated vaccine are potentially superior to killed vaccines because they.

Induce humoral immunity along with CMI.

Are more effective after single dose administration.

Can be given orally with no risk of prick, pain and needle contamination.

Induce immune response at various mucosal sites.

Are cost-effective, easily stored without the need for cold chain, and can be lyophilised.

Understanding of Salmonella genetics, growth and survival in the host are very helpful for developing a rational attenuated vaccine. Attenuation should produce an organism with diminished ability to grow inside the host. Attenuated strain persists in the host to induce protective immunity than the killed vaccine and their growth should be limited even in the immune-deficient hosts. Various genes of Salmonella are already identified for virulence and survival. These genes either belong to housekeeping such as bacterial structural components (LPS, OMP), synthesis of essential metabolites such as purine or pyrimidine biosynthesis or virulence gene involved in bacterial resistance to host defence system [54]. Live attenuated oral vaccines have the good potential to activate lymphocyte expression of the mucosal receptor. Live attenuated vaccine of Salmonella expresses their all antigens which induce broad-spectrum immunity to protect the host from Salmonella serovars. The optimal level of attenuation which does not affect the expression of immunogenicity is a major problem in developing a live attenuated vaccine. We need a live attenuated vaccine which is heat stable and requires few doses. Currently the following attenuated vaccines against typhoid are under different phases of trials.

CVD 909

Live attenuated CVD909 is triple gene mutant of aroC, aroD and htrA along with replacement of PtviA promoter (regulator of Vi antigen expression) with constitutive Ptac (recombinant promoter made by fusion of lac promoter with trp promoter) promoter. Ptac promoter with Vi genes leads to strong and constitutive expression of Vi polysaccharide. The PtviA promoter has an inhibitory effect on Vi expression. This candidate vaccine has the advantage of constitutive expression of Vi antigen which is generally not expressed in Ty21a or inhibited expression or switching off Vi gene expression in the host [55,56].

Ty800

The phoQ is a membrane associated sensor kinase which senses the external environment and induces signaling activation by phosphorylation of PhoP which is a cytoplasmic transcriptional regulator. After induction of PhoP, it can activate and deactivate several genes to combat unfavorable condition. This two-component system is activated inside host macrophage and protect Salmonella from phagocytic activity and from antimicrobial peptides which are secreted by host macrophages for their survival [57-59] The PhoP is crucial for survival inside the mononuclear cell and their mutants in Salmonella typhimurium is highly attenuated and induce immune response in mice but hypersensitive to the microbicidal defensins [60,61]. The live attenuated vaccine of S. typhi is developed for oral administration by mutating global regulator phoP-phoQ gene. This vaccine is based on parent Ty2 strain and developed by Avant Immunotherapeutics. This candidate vaccine has good immunogenicity to induce mucosal and systemic antibodies. The phoP/Q mutants are avirulent inside macrophages. The phoP mutant or constitutive expression of phoP gene was attenuated in mice because it is able to establish infection in Peyer’s patches but unable to infect or reaching spleen. Thus, these mutants are highly suitable for induction of CMI measured by DTH and may be used as an oral candidate vaccine [59-61]. The phoP/Q mutant of Salmonella typhi is safe and immunogenic in humans [62].

clpPX

In Salmonella flhD/flhC is a transcriptional regulator of flagellar gene synthesis. This regulator is degraded by clpPX gene which encodes a protein digesting enzyme. If the clpPX gene is mutated, flagellar gene synthesis is enhanced as the inhibitory action of clpPX is lost. However, clpPX mutated CVD strain of Salmonella is under vaccine trial and outcome is awaited [63].

htrA mutant

The htrA gene encodes heat stress protein of E.coli, and their mutants are avirulent in mice model. HtrA is a serine protease which can cleave misfolded protein in response to heat shock. This protease is induced in heat shock condition and mostly digests periplasmic proteins which are misfolded during the heat stress. The htrA mutant is not heat sensitive but their susceptibility to oxidative stress is increased in macrophages as compared to wild type strain. It protects the mice from re-infection [64]. These gene mutant are safe in normal mice as well as immune-compromised mice such as x-ray irradiated, anti TNFα treated (XID) etc., but it showed some virulence in T-cell deficient mice [65].

ssaV

It is a part of type III secretion system (T3SS) which are encoded by SPI-2. It is required by both the serovars of Typhi and Typhimurium for virulence in human and mice models [66]. SPI-2 encoded genes playing major role in survival of the bacteria inside macrophages of the host [67]. The SsaV protein makes a part of injectosome (T3SS) which secretes various effector proteins outside as well as inside of cell membrane of the host. The ssaV gene mutants are thus made not to secrete effectors protein inside the host cells to cause the disease.

Aromatic Amino Acid Biosynthetic Pathway

Aromatic amino acid biosynthetic genes are needed for bacterial growth. In general three genes are involved in aromatic amino acid biosynthesis viz., aroA, aroC and aroD. Aromatic amino acid biosynthesis gene mutant is a type of auxotrophic mutant which needs a specific metabolite for their growth. Human host is deficient in such metabolites. Therefore, the strain containing these mutations is attenuated in the absence of metabolite and cannot grow in vivo. The bacteria are easily killed but can induce an immune response. If one or more genes were mutated in this operon, it causes irreversible loss of virulence and it also transform from prototroph to auxotroph for Dihydrobenzoate (DHB) and Para-aminobenzoic acid (PABA) [68]. These mutant strains were able to induce an immune response and also protect from the lethal dose of bacterial challenge [69]. These mutants were able to induce both types of immunity viz., cellular as well as humoral immunity [68]. More than one mutation in aro operon (Aromatic amino acid biosynthesis) ensures lack of virulence reversion in bacteria but it does not affect immune inducing potential. A double mutant of aroC and aroD were also used in human volunteers and they showed good immune response but reported causing bacteraemia in some cases [70].

cya (Adenylate cyclase) and crp (cyclic AMP receptor protein)

The cya gene encodes adenylate cyclase enzyme which synthesises cAMP from ATP for various cellular activities. Mostly cAMP is needed for transport of various carbohydrates and amino acids. It also plays a role in the synthesis of flagella, fimbriae and outer membrane protein of bacteria [70]. Oral or intraperitoneal vaccination of single mutant cya or crp gene or double mutant of cya and crp were highly attenuated. Oral vaccine provides protection from oral challenge with wild type strains. When given orally, mutant bacteria invade the Peyer’s patches and mesenteric lymph nodes but unable to invade and survive in spleen in mouse models [71]. In humans, it has been reported that it induces immune response but also gives some side-effects such as fever and bacteraemia.

DNA Vaccines

Injection of naked DNA induces both humoral immunity and CMI. In fact bacterial DNA encodes such a gene which after expression induces Th1 and Th2 responses. For DNA vaccine against typhoid, five peptides were screened that showed high expression in vivo. Those genes are mig14, iicA, sseB, ssaJ, or sifB. Of these five, sseB and mig14 have been observed to be exceptionally efficacious antigens providing specific immunity and protection as compared to other antigens used. Other than high expression level, there are certain antigenic parameters which can influence protective efficacy and show the different immune response for different antigen [72]. For intracellular pathogens, expression of a selected antigen during infection may be more important for the candidate vaccines [Table/Fig-2]. Other vaccine candidate against iNTS (Non Typhoidal Salmonella) and Salmonella Paratyphi A are explained in [Table/Fig-2] [50,51,72-74].

A glimpse of developing vaccines against Salmonellaserotypes [50,51,72-74].

NameSummaryStage of developmentDeveloperReferences
1.Vaccine against Salmonella Paratyphi A
a.O:2-TTO:2 ConjugatePhase 2 and under clinical trial-Technology transfer from NIH to Chengdu Institute (China).-Changchun Institute of Biological Products.Konadu E et al., 1996 [50]
b.O:2-CRM197O:2 ConjugatePreclinicalNVGH (Novartis Vaccine Institute for Global Health)Micoli F, et al., 2012 [51]
2.Vaccine against iNTS
a.O:4,5/O:9-CRMO:4,5/O:9 ConjugatePreclinicalNVGH (Novartis Vaccine Institute for Global Health)Micoli F et al., 2012 [51]
b.WT05Live attenuatedPhase 1Microscience, WokinghamBerkshireHindle Z, et al., 2002 [73]
c.Salmonella hfq mutantmutant Live attenuatedPreclinicalIndian Institute of Science,BangaloreAllam US, et al., 2011 [74]
d.DNA vaccineAroQ, IicA, Mig-14, SsaJ, SsaV, SseB, SifA, SifB, Stm4065, and VirKMice StudyMax Planck Institute for Infection Biology, GermanyRollenhagen C, et al., 2004 [72]

Different types of attenuated new candidates are under research such as recA, recB, dam, waaN, cdt, rpoS etc [Table/Fig-3]. All the other new targets for vaccine development are explained in [Table/Fig-3] [75-82].

List of new targets for vaccine development [75-82].

S. No.GeneFunctionProtection in model animalReference
1relA, spoTPlay a role in stringent responseProtective in murine modelNa HS et al., 2006 [75]
2.damDNA adenine methylase (it controls virulence genes)Protective in murine modelHeithoff DM et al, 2001 [76]
3.recA, recBCDNA recombination and repairNABuchmier NA et al., 1993 [77]
4.cdtColonisation as deep regulatorNAZhang X et al., 1997 [78]
5.wecAUDP-N acetylglucosamine 1-phosphate transferasepersistent infection provide protection from lethal challengeGilbreath JJ et al., 2012 [79]
6.gidAGlucose inhibited divisionInduce immunityShippy DC et al., 2012 [80]
7.rpoSAlternative sigma factor for stress responseImmune to lethal infectionCoynault C et al., 1996 [81]
8.surABiosynthesis of a peptidylprolyl-cis, trans-isomeraseImmune to virulent strain in mice modelSydenham M et al., 2000 [82]

Conclusion

Despite the currently available two vaccines (Ty21a and Vi) Salmonella serotype Typhi mediated problem is still existing in developing countries and causing significant morbidity and mortality. The search of an ideal vaccine against human restricted Salmonella serotypes viz., Typhi and Paratyphi-A, B, C is going on. There is a need to develop satisfactorily attenuated and multivalent subunit candidate vaccine against different serotypes and capable of inducing cell mediated, long lasting immune response. The issue of an efficacious and potent vaccine against typhoid fever inducing strong humoral as well as cellular immunity which can be given to children below 2 years of age still remains to be solved.

References

[1]World Health Organization. Background paper to SAGE on Typhoid Policy Recommendations. 2017  [Google Scholar]

[2]Sinha A, Sazawal S, Kumar R, Sood S, Reddaiah VP, Singh B, Typhoid fever in children aged less than 5 years Lancet 1999 354:734-37.10.1016/S0140-6736(98)09001-1  [Google Scholar]  [CrossRef]

[3]Gonzalez-Escobedo G, Marshall JM, Gunn JS, Chronic and acute infection of the gall bladder by Salmonella Typhi: Understanding the carrier state Nat Rev Microbiol 2011 9(1):09-14.10.1038/nrmicro249021113180  [Google Scholar]  [CrossRef]  [PubMed]

[4]Scanu T, Spaapen RM, Bakker JM, Pratap CB, Wu L, Ingrid H, Salmonella manipulation of host signaling pathways provokes cellular transformation associated with gallbladder carcinoma Cell Host & Microbe 2015 17(6):763-77.10.1016/j.chom.2015.05.00226028364  [Google Scholar]  [CrossRef]  [PubMed]

[5]Ahirwar SK, Pratap CB, Patel SK, Shukla VK, Singh IG, Nath G, Acid exposure induces multiplication of Salmonella enteric Serovar Typhi J Clin Microbiol 2014 52(12):4330-33.10.1128/JCM.02275-1425320227  [Google Scholar]  [CrossRef]  [PubMed]

[6]Carter PB, Collins FM, The route of enteric infection in normal mice Journal of Exp Med 1974 139:118920310.1084/jem.139.5.11894596512  [Google Scholar]  [CrossRef]  [PubMed]

[7]Mirza SH, Beeching NJ, Hart CA, Multi-drug resistant typhoid: A global problem J Med Microbiol 1996 44:317-19.10.1099/00222615-44-5-3178636944  [Google Scholar]  [CrossRef]  [PubMed]

[8]Threlfall JE, Ward LR, Decreased susceptibility to ciprofloxacin in Salmonella enterica serotype Typhi, United Kingdom Emerg Infect Dis 2001 7:448-50.10.3201/eid0703.017315  [Google Scholar]  [CrossRef]

[9]Nath G, Maurya P, Drug resistance patterns in Salmonella enterica subspecies enterica serotype Typhi strains isolated over a period of two decades, with special reference to ciprofloxacin and ceftriaxone Int J Antimicrob Agents 2010 35(5):482-85.10.1016/j.ijantimicag.2009.12.02020188522  [Google Scholar]  [CrossRef]  [PubMed]

[10]Patel SR, Bharti S, Pratap CB, Nath G, Drug resistance pattern in the recent isolates of Salmonella Typhi with special reference to cephalosporins and azithromycin in the gangetic plain JCDR 2017 11(6):DM01-DM03.10.7860/JCDR/2017/23330.997328764168  [Google Scholar]  [CrossRef]  [PubMed]

[11]Saxen H, Reima I, Makela PH, Alternative complement pathway activation by Salmonella O polysaccharide as a virulence determinant in the mouse Microb Pathog 1987 2:15-28.10.1016/0882-4010(87)90111-2  [Google Scholar]  [CrossRef]

[12]Crawford RW, Rosales-Reyes R, Ramırez-Aguilar M de la, Chapa-Azuela O, Alpuche-Aranda C, Gunn JS, Gallstones play a significant role in Salmonella spp. gallbladder colonization and carriage Proc Natl Acad Sci USA 2010 107:4353-58.10.1073/pnas.100086210720176950  [Google Scholar]  [CrossRef]  [PubMed]

[13]Butler T, Ho M, Acharya G, Tiwari M, Gallati H, Interleukin-6, gamma interferon, and tumor necrosis factor receptors in typhoid fever related to outcome of antimicrobial therapy Antimicrob Agents Chemother 1993 37(11):2418-21.10.1128/AAC.37.11.24188285627  [Google Scholar]  [CrossRef]  [PubMed]

[14]Thompson LJ, Dunstan SJ, Dolecek C, Perkins T, House D, Dougan G, Transcriptional response in the peripheral blood of patients infected with Salmonella enterica serovar Typhi Proc Natl Acad Sci USA 2009 106(52):22433-38.10.1073/pnas.091238610620018727  [Google Scholar]  [CrossRef]  [PubMed]

[15]Pietilä TE, Veckman V, Kyllönen P, Lähteenmäki K, Korhonen TK, Julkunen Activation, cytokine production, and intracellular survival of bacteria in Salmonella-infected human monocyte-derived macrophages and dendritic cells J Leukoc Bio 2005 78(4):909-20.10.1189/jlb.120472116033811  [Google Scholar]  [CrossRef]  [PubMed]

[16]Näsström E, Parry CM, Vu Thieu NT, Maude RR, de Jong HK, Fukushima M, Reproducible diagnostic metabolites in plasma from typhoid fever patients in Asia and Africa eLife 2017 6:e1565110.7554/eLife.1565128483042  [Google Scholar]  [CrossRef]  [PubMed]

[17]Vidal SM, Malo D, Vogan K, Skamene E, Gros P, Natural resistance to infection with intracellular parasites: Isolation of a candidate for BCG Cell 1993 73:469-86.10.1016/0092-8674(93)90135-D  [Google Scholar]  [CrossRef]

[18]Hess J, Ladel C, Miko D, Kaufmann SH, Salmonella Typhimurium aroA- infection in gene-targeted immunodeficient mice: major role of CD4+TCR-alpha beta cells and IFN-gamma in bacterial clearance independent of intracellular location J Immunol 1996 156(9):3321-26.  [Google Scholar]

[19]McSorley SJ, Jenkins MK, Antibody is required for protection against virulent but not attenuated Salmonella enterica serovar Typhimurium Infect Immun 2000 68:3344-48.10.1128/IAI.68.6.3344-3348.2000  [Google Scholar]  [CrossRef]

[20]Mastroeni P, Villarreal-Ramos B, Hormaeche CE, Adoptive transfer of immunity to oral challenge with virulent salmonellae in innately susceptible BALB/c mice requires both immune serum and T cells Infect Immun 1993 61(9):3981-84.  [Google Scholar]

[21]Harrison JA, Villarreal-Ramos B, Mastroeni P, Demarco HR, Hormaeche CE, Correlates of protection induced by live Aro- Salmonella Typhimurium vaccines in the murine typhoid model Immunology 1997 90:618-25.10.1046/j.1365-2567.1997.00158.x9176117  [Google Scholar]  [CrossRef]  [PubMed]

[22]Svenson SB, Lindberg AA, Artificial Salmonella vaccines: Salmonella Typhimurium O-antigen specific oligosaccharide-protein conjugates elicit protective antibodies in rabbits and mice Infect Immun 1981 32(2):490-96.  [Google Scholar]

[23]Kossaczka Z, Lin FY, Ho VA, Thuy NT, Van bay P, Tanh TC, Safety and immunogenicity of Vi conjugate vaccines for typhoid fever in adults, teenagers, and 2- to 4-year-old children in Vietnam Infect Immun 1999 67:5806-10.  [Google Scholar]

[24]Kumar VU, Muthukkaruppan VR, An outer membrane protein (porin) as an eliciting antigen for delayed-type hypersensitivity in murine salmonellosis Infect Immun 1987 55:822-24.  [Google Scholar]

[25]Fraser A, Paul M, Goldberg E, Acosta CJ, Leibovici L, Typhoid fever vaccines: Systematic review and metaanalysis of randomised controlled trials Vaccine 2007 25:7848-57.10.1016/j.vaccine.2007.08.02717928109  [Google Scholar]  [CrossRef]  [PubMed]

[26]Khan MI, Ochiai RL, Clemens JD, Population impact of Vi capsular polysaccharide vaccine Expert Rev Vaccines 2010 9:485-96.10.1586/erv.10.4320450323  [Google Scholar]  [CrossRef]  [PubMed]

[27]Wong KH, Feeley JC, Northrup RS, Forlines ME, Vi antigen from Salmonella typhosa and immunity against typhoid fever. I. Isolation and immunologic properties in animals Infect Immun 1974 9:348-53.  [Google Scholar]

[28]Garmory HS, Brown KA, Titball RW, Salmonella vaccines for use in humans: Present and future perspectives FEMS Microbiol Rev 2002 26:339-53.10.1111/j.1574-6976.2002.tb00619.x12413664  [Google Scholar]  [CrossRef]  [PubMed]

[29]Galdiero M, De martino L, Marcatili A, Nuzzo I, Vitiello IM, Cipollardo de lero G, Th1 and Th2 cell involvement in immune response to Salmonella Typhimurium porins Immunology 1998 94(1):05-13.10.1046/j.1365-2567.1998.00492.x9708180  [Google Scholar]  [CrossRef]  [PubMed]

[30]Felix A, Krikorian KS, Reitler R, The occurrence of typhoid bacilli containing Vi antigen in cases of typhoid fever and of Vi antibody in their sera J Hyg 1935 35:421-27.10.1017/S002217240003244720475293  [Google Scholar]  [CrossRef]  [PubMed]

[31]Hessel L, Debois H, Fletcher M, Dumas R, Experience with Salmonella Typhi Vi capsular polysaccharide vaccine Eur J Clin Microbiol Infect Dis 1999 18:609-20.10.1007/s10096005036110534182  [Google Scholar]  [CrossRef]  [PubMed]

[32]Yang HH, Wu CG, Xie GZ, Gu QW, Wang BR, Wang LY, Efficacy trial of Vi polysaccharide vaccine against typhoid fever in southwestern China Bull World Health Organ 2001 79:625-31.  [Google Scholar]

[33]Saha MR, Ramamurthy T, Dutta P, Mitra U, Emergence of Salmonella Typhi Vi antigen negative strains in an epidemic of multidrug resistant typhoid fever cases in Calcutta, India Natl Med J India 2000 13:164  [Google Scholar]

[34]Singh M, Ganguly NK, Kumar L, Vohra H, Protective efficacy and immunogenicity of Vi porin conjugate against Salmonella Typhi Microbiology Immuno 1999 43:535-42.10.1111/j.1348-0421.1999.tb02439.x10480549  [Google Scholar]  [CrossRef]  [PubMed]

[35]Ivanoff B, Levine MM, Lambert PH, Vaccination against typhoid fever: present status Bull World Health Organ 1994 72:957-71.  [Google Scholar]

[36]World Health Organization. Diarrhoeal diseases. Geneva: WHO; 2009  [Google Scholar]

[37]Voysey M, Pollard AJ, Sero-efficacy of Vi-polysaccharide tetanus-toxoid typhoid conjugate vaccine (Typbar-TCV) Clin Infect Dis 2018 67(1):18-24.10.1093/cid/cix114529351594  [Google Scholar]  [CrossRef]  [PubMed]

[38]Poolman J, Borrow R, Hyporesponsiveness and its clinical implications after vaccination with polysaccharide or glycoconjugate vaccines Expert Rev Vaccines 2011 10(3):307-22.10.1586/erv.11.821434799  [Google Scholar]  [CrossRef]  [PubMed]

[39]Kantele A, Pakkanen SH, Siitonen A, Karttunen R, Kantele JM, Live oral typhoid vaccine Salmonella Typhi Ty21a-a surrogate vaccine against nontyphoid Salmonella? Vaccine 2012 30:7238-45.10.1016/j.vaccine.2012.10.00223084770  [Google Scholar]  [CrossRef]  [PubMed]

[40]Sur D, Ochiai RL, Bhattacharya SK, Ganguly NK, Ali M, Manna B, A cluster-randomized effectiveness trial of Vi typhoid vaccine in India N Engl J Med 2009 361:335-44.10.1056/NEJMoa080752119625715  [Google Scholar]  [CrossRef]  [PubMed]

[41]Mai NL, Phan VB, Vo AH, Tran CT, Lin FY, Bryla DA, Persistent efficacy of Vi conjugate vaccine against typhoid fever in young children N Engl J Med 2003 349:1390-91.10.1056/NEJM20031002349142314523155  [Google Scholar]  [CrossRef]  [PubMed]

[42]Thiem VD, Lin FY, Canh G, Son NH, Anh DD, Mao ND, The Vi conjugate typhoid vaccine is safe, elicits protective levels of IgG anti-Vi, and is compatible with routine infant vaccines Clin Vaccine Immunol 2011 18:730-35.10.1128/CVI.00532-1021411598  [Google Scholar]  [CrossRef]  [PubMed]

[43]Szu SC, Developments of Vi conjugate- A new generation of typhoid vaccine Expert Rev Vaccines 2013 12:1273-86.10.1586/14760584.2013.84552924156285  [Google Scholar]  [CrossRef]  [PubMed]

[44]Sette A, Rappuoli R, Reverse vaccinology: Developing vaccines in the era of genomics Immunity 2010 33:530-41.10.1016/j.immuni.2010.09.01721029963  [Google Scholar]  [CrossRef]  [PubMed]

[45]Bobat S, Flores-Langarica A, Hitchcock J, Marshall JL, Kingsley RA, Goodall M, Soluble flagellin, FliC, induces an Ag-specific Th2 response, yet promotes T-bet-regulated Th1 clearance of Salmonella Typhimurium infection Eur J Immunol 2011 41:1606-18.10.1002/eji.20104108921469112  [Google Scholar]  [CrossRef]  [PubMed]

[46]Simon R, Levine MM, Glycoconjugate vaccine strategies for protection against invasive Salmonella infections Hum Vaccin Immunother 2012 8:494-98.10.4161/hv.1915822370510  [Google Scholar]  [CrossRef]  [PubMed]

[47]Van Damme P, Kafeja F, Anemona A, Basile V, Hilbert AK, De Coster I, Safety, immunogenicity and dose ranging of a new Vi-CRM197 conjugate vaccine against typhoid fever: Randomized clinical testing in healthy adults PLoS One 2011 6:e2539810.1371/journal.pone.002539821980445  [Google Scholar]  [CrossRef]  [PubMed]

[48]Bhutta ZA, Capeding MR, Bavdekar A, Marchetti E, Ariff S, Soofi SB, Immunogenicity and safety of the Vi-CRM197 conjugate vaccine against typhoid fever in adults, children, and infants in South and South East Asia: results from two randomized, observer-blind, age de-escalation, phase 2 trials Lancet Infect Dis 2014 14:119-29.10.1016/S1473-3099(13)70241-X  [Google Scholar]  [CrossRef]

[49]Cui C, Carbis R, An SJ, Jang H, Czerkinsky C, Szu SC, Physical and chemical characterization and immunologic properties of Salmonella enterica serovar Typhi capsular polysaccharide diphtheria toxoid conjugates Clin Vaccine Immunol 2010 17:73-79.10.1128/CVI.00266-0919889941  [Google Scholar]  [CrossRef]  [PubMed]

[50]Konadu E, Shiloach J, Bryla DA, Robbins JB, Szu SC, Synthesis, characterization, and immunological properties in mice of conjugates composed of detoxified lipopolysaccharide of Salmonella Paratyphi A bound to tetanus toxoid with emphasis on the role of O acetyls Infect Immun 1996 64:2709-15.  [Google Scholar]

[51]Micoli F, Rondini S, Gavini M, Lanzilao L, Medaglini D, Saul A, O:2-CRM(197) conjugates against Salmonella Paratyphi A PLoS One 2012 7:e4703910.1371/journal.pone.004703923144798  [Google Scholar]  [CrossRef]  [PubMed]

[52]Ni Y, Springer MJ, Guo J, Finger-Baker I, Wilson JP, Cobb RR, Development of a synthetic Vi polysaccharide vaccine for typhoid fever Vaccine 2017 35(51):7121-26.10.1016/j.vaccine.2017.10.08129150208  [Google Scholar]  [CrossRef]  [PubMed]

[53]Datsenko KA, Kirill A, Wanner BL, One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products Proc Natl Acad Sci USA 2000 97(12):6640-45.10.1073/pnas.12016329710829079  [Google Scholar]  [CrossRef]  [PubMed]

[54]FieldsI PI, Swanson RV, Haidaris CG, Heffron F, Mutants of Salmonella Typhimurium that cannot survive within the macrophage are avirulent Proc Natl Acad Sci USA 1986 83:5189-93.10.1073/pnas.83.14.51893523484  [Google Scholar]  [CrossRef]  [PubMed]

[55]Wang JY, Noriega FR, Galen JE, Barry E, Levine MM, Constitutive expression of the Vi polysaccharide capsular antigen in attenuated Salmonella enterica serovar Typhi oral vaccine strain CVD 909 Infect Immun 2000 68:4647-52.10.1128/IAI.68.8.4647-4652.200010899868  [Google Scholar]  [CrossRef]  [PubMed]

[56]Wahid R, Salerno-Gonçalves R, Tacket CO, Levine MM, Sztein MB, Cell-mediated immune responses in humans after immunization with one or two doses of oral live attenuated typhoid vaccine CVD909 Vaccine 2007 25:1416-25.10.1016/j.vaccine.2006.10.04017182155  [Google Scholar]  [CrossRef]  [PubMed]

[57]Groisman EA, Chiao E, Lipps CJ, Heffron F, Salmonella Typhimurium phoP virulence gene is a transcriptional regulator Proc Natl Acad Sci USA 1989 86(18):7077-81.10.1073/pnas.86.18.70772674945  [Google Scholar]  [CrossRef]  [PubMed]

[58]Belden WJ, Miller SI, Further characterization of the PhoP regulon: Identification of new PhoP-activated virulence loci Infect Immun 1994 62(11):5095-101.  [Google Scholar]

[59]Galan JE, Curtiss RD, Virulence and vaccine potential of phoP mutants of Salmonella Typhimurium Microbial Pathog 1989 6:433-43.10.1016/0882-4010(89)90085-5  [Google Scholar]  [CrossRef]

[60]Groisman EA, Saier MH JR, Salmonella virulence: New clues to intramacrophage survival Trends in Biochemi Sci 1990 15:30-33.10.1016/0968-0004(90)90128-X  [Google Scholar]  [CrossRef]

[61]Miller SI, PhoP/PhoQ: macrophage-specific modulators of Salmonella virulence? Mol Micro 1991 5:2073-78.10.1111/j.1365-2958.1991.tb02135.x1766380  [Google Scholar]  [CrossRef]  [PubMed]

[62]Hohmann EL, Oletta CA, Killeen KP, Miller SI, PhoP/phoQ deleted Salmonella Typhi (Ty800) is a safe and immunogenic single-dose typhoid fever vaccine in volunteers J Infect Dis 1996 173:1408-14.10.1093/infdis/173.6.14088648213  [Google Scholar]  [CrossRef]  [PubMed]

[63]Tomoyasu T, Ohkishi T, Ukyo Y, Tokumitsu A, Takaya A, Suzuki M, The ClpXP ATP-dependent protease regulates flagellum synthesis in Salmonella enterica serovar Typhimurium J Bacteriol 2002 184:645-53.10.1128/JB.184.3.645-653.200211790733  [Google Scholar]  [CrossRef]  [PubMed]

[64]Pallen MJ, Wren BW, The HtrA family of serine proteases Mol Micro 1997 26:209-21.10.1046/j.1365-2958.1997.5601928.x9383148  [Google Scholar]  [CrossRef]  [PubMed]

[65]Strahan K, Chatfield SN, Tite J, Dougan G, Hormaeche CE, Impaired resistance to infection does not increase the virulence of Salmonella htrA live vaccines for mice Microbial Pathog 1992 12:311-7.10.1016/0882-4010(92)90049-T  [Google Scholar]  [CrossRef]

[66]Shea JE, Hensel M, Gleeson C, Holden DW, Identification of a virulence locus encoding a second type III secretion system in Salmonella Typhimurium Proc Natl Acad Sci USA 1996 93:2593-97.10.1073/pnas.93.6.25938637919  [Google Scholar]  [CrossRef]  [PubMed]

[67]Ochman H, Soncini FC, Solomon F, Groisman EA, Identification of a pathogenicity island required for Salmonella survival in host cells Proc Nat Acad Sci USA 1996 93:7800-04.10.1073/pnas.93.15.78008755556  [Google Scholar]  [CrossRef]  [PubMed]

[68]Hoiseth SK, Stocker BA, Aromatic-dependent Salmonella Typhimurium are non virulent and effective as live vaccines Nature 1981 291:238-39.10.1038/291238a07015147  [Google Scholar]  [CrossRef]  [PubMed]

[69]Hormaeche CE, Mastroeni P, Harrison JA, Demarco de Hormaeche R, venson S, Protection against oral challenge three months after i.v. immunization of BALB/c mice with live Aro Salmonella typhimurium and Salmonella enteritidis vaccines is serotype (species)-dependent and only partially deter-mined by the main LPS-O antigen Vaccine 1996 14:251-59.10.1016/0264-410X(95)00249-Z  [Google Scholar]  [CrossRef]

[70]Tacket CO, Hone DM, Curtiss RD, Kelly SM, Losonsky G, Guers L, Comparison of the safety and immunogenicity of ΔaroC ΔaroD and Δcya Δcrp Salmonella Typhi strains in adult volunteers Infect Immun 1992 60:536-41.  [Google Scholar]

[71]Curtiss RD, Kelly SM, Salmonella Typhimurium deletion mutants lacking adenylate cyclase and cyclic AMP receptor protein are avirulent and immunogenic Infect Immun 1987 55:3035-43.  [Google Scholar]

[72]Rollenhagen C, Sorensen M, Rizos K, Hurvitz R, Bumann D, Antigen selection based on expression levels during infection facilitates vaccine development for an intracellular pathogen Proc Natl Acad Sci USA 2004 101:8739-44.10.1073/pnas.040128310115173591   [Google Scholar]  [CrossRef]  [PubMed]

[73]Hindle Z, Chatfield SN, Phillimore J, Bentley M, Johnson J, Cosgrove CA, Characterization of Salmonella enterica derivatives harboring defined aroC and Salmonella pathogenicity island 2 type III secretion system (ssaV) mutations by immunization of healthy volunteers Infect Immun 2002 70:3457-67.10.1128/IAI.70.7.3457-3467.200212065485  [Google Scholar]  [CrossRef]  [PubMed]

[74]Allam US, Krishna MG, Lahiri A, Joy O, Chakravortty D, Salmonella enterica serovar Typhimurium lacking hfq gene confers protective immunity against murine typhoid PLoS One 2011 6:e1666710.1371/journal.pone.001666721347426  [Google Scholar]  [CrossRef]  [PubMed]

[75]Na HS, Kim HJ, Lee HC, Hong Y, Rhee JH, Choy HE, Immune response induced by Salmonella Typhimurium defective in ppGpp synthesis Vaccine 2006 24:2027-34.10.1016/j.vaccine.2005.11.03116356600  [Google Scholar]  [CrossRef]  [PubMed]

[76]Heithoff DM, Enioutina EY, Daynes RA, Sinsheimer RL, Low DA, Mahan MJ, Salmonella DNA adenine methylase mutants confer cross-protective immunity Infect Immun 2001 69:6725-30.10.1128/IAI.69.11.6725-6730.200111598044  [Google Scholar]  [CrossRef]  [PubMed]

[77]Buchmeier NA, Lipps CJ, So MY, Heffron F, Recombination-deficient mutants of Salmonella Typhimurium are avirulent and sensitive to the oxidative burst of macrophages Mol Micro 1993 7:933-36.10.1111/j.1365-2958.1993.tb01184.x8387147  [Google Scholar]  [CrossRef]  [PubMed]

[78]Zhang X, Kelly SM, Bollen WS, Curtiss 3rd R, Characterization and immuno-genicity of Salmonella Typhimurium SL1344 and UK-1 delta crp and delta cdt deletion mutants Infect Immun 1997 65:5381-87.  [Google Scholar]

[79]Gilbreath JJ, Colvocoresses Dodds J, Rick PD, Soloski MJ, Merrell DS, Metcalf ES, Enterobacterial common antigen mutants of Salmonella enterica serovar Typhimurium establish a persistent infection and provide protection against subsequent lethal challenge Infect Immun 2012 80:441-50.10.1128/IAI.05559-1122025511  [Google Scholar]  [CrossRef]  [PubMed]

[80]Shippy DC, Fadl AA, Immunological characterization of a gidA mutant strain of Salmonella for potential use in a live-attenuated vaccine BMC Microbiol 2012 12:28610.1186/1471-2180-12-28623194372  [Google Scholar]  [CrossRef]  [PubMed]

[81]Coynault C, Robbe-Saule V, Norel F, Virulence and vaccine potential of Salmonella Typhimurium mutants deficient in the expression of the RpoS (sigma S) regulon Mol Micro 1996 22:149-60.10.1111/j.1365-2958.1996.tb02664.x8899717  [Google Scholar]  [CrossRef]  [PubMed]

[82]Sydenham M, Douce G, Bowe F, Ahmed S, Chatfield S, Dougan G, Salmonella enterica serovar Typhimurium surA mutants are attenuated and effective live oral vaccines Infect Immun 2000 68:1109-15.10.1128/IAI.68.3.1109-1115.200010678914  [Google Scholar]  [CrossRef]  [PubMed]