Skip to main content
Taylor & Francis Open Select logoLink to Taylor & Francis Open Select
. 2016 Jan 23;42(6):928–941. doi: 10.3109/1040841X.2015.1105782

Is gonococcal disease preventable? The importance of understanding immunity and pathogenesis in vaccine development

Jennifer L Edwards a,*, Michael P Jennings b, Michael A Apicella c, Kate L Seib b,*
PMCID: PMC4958600  NIHMSID: NIHMS787842  PMID: 26805040

Abstract

Gonorrhea is a major, global public health problem for which there is no vaccine. The continuing emergence of antibiotic-resistant strains raises concerns that untreatable Neisseria gonorrhoeae may become widespread in the near future. Consequently, there is an urgent need for increased efforts towards the development of new anti-gonococcal therapeutics and vaccines, as well as suitable models for potential pre-clinical vaccine trials. Several current issues regarding gonorrhea are discussed herein, including the global burden of disease, the emergence of antibiotic-resistance, the status of vaccine development and, in particular, a focus on the model systems available to evaluate drug and vaccine candidates. Finally, alternative approaches to evaluate vaccine candidates are presented. Such approaches may provide valuable insights into the protective mechanisms, and correlates of protection, required to prevent gonococcal transmission, local infection and disease sequelae.

Keywords: Candidate vaccine antigens, gonorrhea, infection models, multi-drug resistance, Neisseria gonorrhoeae

Introduction

Infections caused by Neisseria gonorrhoeae (the gonococcus) continue to be a global, intractable problem. The absence of a gonococcal vaccine, together with the continuing emergence of antibiotic-resistant and untreatable gonococcal strains, has raised awareness that N. gonorrhoeae poses an “urgent” public health threat for which immediate aggressive action is greatly needed (Unemo & Shafer, 2014). Herein, we review the impediments and potential opportunities to gonococcal vaccine development.

The global burden of gonorrhea

The highest burden of N. gonorrhoeae disease occurs in men and women between 15 and 29 years of age (CDC, 2013; WHO, 2012). Although infection typically results in urethritis in males and cervicitis in females, mucosal infections of the rectum, pharynx and eye are also commonly reported (Hook & Handsfield, 2008). The World Health Organization (WHO) reports an estimated global incidence of over 106 million cases per year, with a 21% increase in incidence having occurred between 2005 and 2008 (WHO, 2012). More recent data indicate an 11% increase in the number of reported cases in the United States from 2009 to 2013 (CDC, 2014) and a 90% increase in Australia from 2009 to 2014 (NNDSS, 2015). Incidence is likely underestimated, however, because of inadequate surveillance and diagnostics methods in many regions, as well as the high number of asymptomatic cases. In this regard, if not promptly diagnosed and treated, infection can lead to severe reproductive complications. Neisseria gonorrhoeae disease sequelae are predominately observed among women, owing to the higher proportion of asymptomatic infections in females [50–80% (Farley et al., 2003; Hook & Handsfield, 2008; WHO, 2011)] versus males [reported to be from 1% to 40% (Farley et al., 2003; Handsfield et al., 1974; Hook & Handsfield, 2008; Johnson et al., 2010; WHO, 2011)]. Ascending reproductive tract infections lead to pelvic inflammatory disease (PID, i.e. inflammation of the uterus, fallopian tubes, ovaries, etc.) in 10–40% of infected women that, in turn, can cause infertility and ectopic pregnancies. Additional disease sequelae include fetal wastage, neonatal conjunctivitis, adverse pregnancy outcomes and disseminated gonococcal infections (Hook & Handsfield, 2008). Complications of infection are rare in males, but they include urethral stricture and urogenital tract abscesses, as well as inflammation of the prostate gland, epididymis and/or testes (Hook & Handsfield, 2008). In both sexes, infection with N. gonorrhoeae imposes an additional increased risk to contract and transmit HIV (Chen et al., 2003; Cohen et al., 1997; Levine et al., 1998). In terms of economic burden, gonococcal infections in the United States are estimated to account for annual direct medical costs exceeding $162.1 million (Owusu-Edusei et al., 2013) and indirect costs in excess of $1.1 billion (Aledort et al., 2005).

Current situation for gonococcal control

Presently, the control of N. gonorrhoeae largely relies on prompt diagnosis and antibiotic treatment of infected persons and their acknowledged sex partners (CDC, 2013). However, high levels of asymptomatic infections (as described above) and low levels of routine screening (Huppert et al., 2005) hamper prompt diagnosis, even though available diagnostics are highly sensitive (>90%) and specific (>99%) (Papp et al., 2014). Furthermore, there is a heavy reliance upon syndromic disease management (i.e. the treatment of all at risk patients based upon the presence of symptoms rather than a definitive diagnosis) in many of the countries that are most heavily impacted by gonococcal disease. Syndromic disease management is not suitable for the diagnosis and treatment of the high numbers of individuals that present with asymptomatic disease. Moreover, it contributes to antibiotic-resistance by the overuse and misuse of antibiotics. In this regard, the gonococcus has developed resistance to all classes of antibiotics used to treat it over the past seven decades, including the sulphonamides, penicillins, tetracyclines, macrolides and quinolones (reviewed in Unemo & Shafer, 2014). Multi-drug-resistant strains are of the greatest concern and have been identified in Japan (Ohnishi et al., 2011), Europe (Unemo et al., 2011, 2012), South Africa (Lewis et al., 2013), Australia (Lahra et al., 2014) and North America (Barry & Klausner, 2009; Martin et al., 2012). These strains exhibit high-level resistance to the expanded-spectrum cephalosporins, ceftriaxone and cefixime; which are the last remaining options for empirical first-line treatment. Combined antibiotic therapies are currently being evaluated in clinical trials (Kirkcaldy et al., 2014), but there are no new single antibiotic options presently available, or in the pipeline, and it is of concern that even new classes of antibiotics would only offer a short-term therapeutic solution (Tapsall et al., 2009; Unemo & Nicholas, 2012). An example of this rapid change in antimicrobial-resistance is exemplified by the fact that the 2010 Centers for Disease Control and Prevention (CDC) recommendations for gonorrhea treatment had to be revised in 2012 to include the use of double antibiotic therapy (CDC, 2012). Additionally, diminished effectiveness of cefixime, an oral cephalosporin, has prompted the recommended use of ceftriaxone, which is administered by muscular injection (CDC, 2012). Therefore, more aggressive and invasive treatment is now required to treat N. gonorrhoeae infections. Recognizing infections with antibiotic-resistant strains and, hence preventing treatment failures, is also becoming increasingly difficult because of the general shift from culture-based diagnostic methods (required for susceptibility testing) to nucleic acid amplification testing. Additionally, most national surveillance bodies only monitor antimicrobial-resistance for a small percentage of reported cases [e.g. fewer than 2% of all reported gonorrhea cases are sampled by the USA Gonococcal Isolate Surveillance Project (GISP) (Bolan et al., 2012)].

The CDC has listed N. gonorrhoeae as one of the three microorganisms that pose an urgent threat with regard to antibiotic-resistance – highlighting it as an “immediate public-health threat that requires urgent and aggressive action” (CDC, 2013). This threat assessment also included the evaluation of the clinical and economic impact, incidence, 10-year projection of incidence, transmissibility, availability of effective antibiotics and barriers to prevention for gonorrhea. Projection of these data indicates there will be an additional 75 000 cases of PID, 15 000 cases of epididymitis and 222 HIV infections during a 10-year period in the United States alone, if cephalosporin-resistant N. gonorrhoeae strains become widespread. Furthermore, direct medical costs will more than double, with greater than $200 million in additional budget being required for increased monitoring and case management, as well as for additional courses of antibiotics (CDC, 2013).

Current status of vaccine development

Vaccination is generally considered the best long-term approach to control infectious diseases. However, efforts over the past century to develop a gonococcal vaccine have proven particularly arduous. Only four candidate vaccines have progressed to clinical trials, (1) a whole cell (therapeutic) vaccine tested during the early 1900s (Eyre & Stewart, 1909), (2) a partially autolyzed vaccine tested in 1974 (Greenberg et al., 1974), (3) pilus-based vaccines tested in the 1990s (Boslego et al., 1991) and (4) a protein I-based vaccine derived by differential centrifugation of disrupted gonococci (reviewed in Tramont, 1989). None of these vaccines were successful, despite preliminary evidence to suggest that (1) whole cell (mixed strain) vaccination therapy could improve infection outcomes in patients (Eyre & Stewart, 1909) and (2) human anti-pilus antibodies, and (antibody-containing) genital secretions, could block subsequent in vitro adherence to human buccal epithelial cells (Tramont et al., 1980, 1981). These latter data potentially highlight the importance of the use of appropriate infection models for pre-clinical N. gonorrhoeae vaccine evaluation, in that gonococci do not naturally colonize buccal cells. Moreover, these studies provided the first insight into the impending numerous difficulties in N. gonorrhoeae vaccine development, which are primarily the result of the variability of gonococcal antigens, production of “blocking” antibodies to conserved gonococcal antigens (Rice et al., 1986), a lack of knowledge of what immune response might confer protective immunity and the absence of robust animal models in which to study this obligate human pathogen, as is discussed further below (reviewed in Jerse et al., 2014; Mietzner et al., 2004; Seib & Rappuoli, 2010).

Candidate gonococcal vaccine antigens

The majority of gonococcal antigens studied to date are extremely variable in the amino acid sequence of protein antigens and in the structure of oligosaccharide antigens. This is true both among strains and within a given strain. High levels of natural transformation, antigenic variation and phase variation drive this variation, which is characteristic of the pathogenic Neisseria (Rotman & Seifert, 2014). The impact of these factors on vaccine development is clear from clinical trials in which gonococci expressing multiple variants of the major surface appendages, pilin (Seifert et al., 1994; Swanson et al., 1987), lipooligosaccharide (LOS) (Schneider et al., 1991) and the opacity-associated outer membrane proteins (Opa) (Jerse et al., 1994; Swanson et al., 1988), were recovered from human male volunteers shortly after infection with a predominantly antigenically-homogenous inoculum. However, several other immunogenic proteins are also characterized that could serve as potential vaccine targets resulting from their high level of antigenic conservation and stable expression within and between strains. These targets include: the nitrite reductase, AniA (Shewell et al., 2013); phospholipase D (PLD) (Apicella & Edwards, 2007; Edwards & Apicella, 2006); transferrin-binding proteins, TbpAB (Price et al., 2007); MtrE of the Mtr efflux pump complex (DeRocco and Jerse, unpublished data reported in Jerse & Deal, 2013), the outer membrane porin, PorB (Garvin et al., 2010; Massari et al., 2003), as well as the conserved 2C7 epitope of LOS (Gulati et al., 2013; Ngampasutadol et al., 2006) [Table 1; (reviewed in Jerse et al., 2014; Jerse & Deal, 2013)]. New gonococcal candidate antigens could also be realized through improved vaccine technologies that support genome wide analyses as well as the rational engineering and redesigning of promising targets (as outlined in Rinaudo et al., 2009). Additionally, novel antigen presentation strategies [e.g. outer membrane vesicles (OMVs) that present several target antigens, as used for serogroup B meningococcal vaccines (Holst et al., 2013)] may facilitate gonococcal vaccine development.

Table 1. Potential gonorrhea vaccine antigens.

Name Role Variability Antibodies Effect of candidate-directed antibody References
Colonization
 PLD Phospholipase D, regulator of gonococcal invasion of and survival within cervical epithelia Highly conserved FI Antibodies inhibiting NgPLD decrease adherence to and invasion of primary cervical cells Apicella & Edwards (2007) and Edwards et al. (2003)
 Pilin Major outer membrane protein, mediates adherence to epithelial cells Antigenically variable FI Antibodies to pili block attachment to human cells, but are directed at variable epitopes Rothbard et al. (1985), Siegel et al. (1982), Schoolnik et al. (1983), Tramont et al. (1981), and Virji & Heckels (1984)
 PilQ Outer membrane channel through which pili are extruded Stable expression B* N/A Haghi et al. (2012)
 PorB Major porin, outer membrane pore; involved in gonococcal invasion of cervical cells through CR3, PorB1A mediates epithelial invasion through the SREC-1 receptor Stable expression, two serogroups (PorB1A and PorB1B) B (Cyclic loop peptides); B* N/A Edwards et al. (2002), Rechner et al. (2007), Garvin et al. (2010), and Massari et al. (2003)
 Opa proteins Mediates adherence to immune cells Phase variable, several antigenically distinct Opa proteins per strain B*, M* N/A Callaghan et al. (2011), Cole & Jerse (2009), and de Jonge et al. (2004)
 OmpA Mediates invasion of malignant cervical and endometrial cell lines Stably expressed, highly conserved B N/A Serino et al. (2007)
Nutrient acquisition and metabolism
 TbpA, TbpB Transferrin (Tf) receptor TbpA and TbpB are highly and semiconserved, respectively B, FI Tf required for infection of male volunteers by strains lacking LbpA and LbpB Antibodies in mice block growth in the presence of Tf as a sole iron source Price et al. (2007) and Hobbs et al. (2011)
 LbpA, LpbB Lactoferrin receptor LbpA and LbpB are semiconserved *, not expressed by all gonococcal strains B* N/A Pettersson et al. (2006), Adamiak et al. (2012), Biswas et al. (1999) and Mickelsen et al. (1982)
 TdfJ (ZnuD*) TonB-dependent zinc transporter Conserved, iron induced B* N/A Cornelissen & Hollander (2011) and Stork et al. (2010)
 AniA Nitrite reductase, required for anaerobic growth and biofilm formation Conserved, induced by low O2 tension and the presence of nitrite FI Antiserum to truncated non-glycosylated recombinant protein blocks AniA nitrite reductase activity Clark et al. (1988), Falsetta et al. (2009) and Shewell et al. (2013)
Evasion of innate defenses
 MtrE Surface-exposed channel of the MtrC-MtrD-MtrE and FarA-FarB-MtrE active efflux pumps Stable expression and highly conserved B N/A Jerse & Deal (2013) (DeRocco and Jerse, unpublished data)
 Lst α2,3 sialyltransferase; catalyzes the addition of host-derived sialic acid to the lacto-N-neotetraose moiety of LOS; protects gonococci from complement, non-opsonic uptake by neutrophils, and antimicrobial peptides Conserved, variable levels of activity between strains, repressed by CrgA FI Antibodies reduce sialylation Smith et al. (1995), Shell et al. (2002) and Packiam et al. (2006)
 NspA Factor H binding and serum resistance Stably expressed, highly conserved B, O N/A Li et al. (2011) and Martin et al. (1997)
 PorB PorB binds C4b-binding protein and factor H to down-regulate complement activation at the gonococcal surface Described above      
Other
 2C7 epitope The 2C7 epitope is a common oligosaccharide structure within gonococcal LOS that is recognized by a murine monoclonal antibody, called 2C7 Phase variable but expressed by >95% of isolates B, O, M N/A Gulati et al. (1996, 2012, 2013) and Ngampasutadol et al. (2006)

B, bactericidal; O, opsonic; M, protective in mouse model of infection; FI, function inhibiting.

*Data for N. meningitidis homologue; N/A, not applicable/no data available.

Immune response to gonococcal infection

Traditional vaccine approaches are based on inactivated or attenuated organisms, or purified antigens, and they aim to replicate natural immunity while simultaneously deterring disease (Rappuoli, 2007). However, natural infection with N. gonorrhoeae does not induce a protective immune response (Fox et al., 1999; Hedges et al., 1999; Schmidt et al., 2001; Ross et al., 1995). Indeed, the Scottish essayist and biographer of Samuel Johnson, James Boswell, wrote detailed diary accounts of his 18 episodes of gonococcal urethritis between 1760 and 1790 (Ober, 1970). It is not clear if this suggests natural clearance of the organism may have occurred in the pre-antibiotic era and, thus, would eventually occur today, if treatment were withheld. However, given the high prevalence of asymptomatic and sub-clinical gonococcal disease, it is unknown whether, in the pre-antibiotic era when diagnosis was based on symptomatology, patients did actually clear the infection. Regardless, it is well known among STI clinicians that patients with gonorrhea frequently have recurrent gonococcal infections (Fowler et al., 2010), indicating a lack of natural immunity. Furthermore, experimental gonococcal infections of human volunteers demonstrated that protection was not conferred, even when subjects were challenged six months after the first infection with the identical strain (Schmidt et al., 2001). Such experiences should tell us that, if natural infection does not provide protection against reinfection, vaccine development will require the generation of a non-native immune response. Therefore, a full understanding of gonococcal pathogenesis is needed to identify possible, and novel, approaches to solve the problem. An increased understanding of the impact of the female genital tract microbiome and co-infection with other STIs on the immune response would also be valuable, as both these factors may alter transmission and disease progression of N. gonorrhoeae (Gallo et al., 2012; Saigh et al., 1978).

The observations that gonococcal infections are often persistent and that reinfection is common are consistent with a growing body of literature indicating that N. gonorrhoeae is able to avoid (Kilian & Reinholdt, 1987; Lewis et al., 2010; Mandrell & Apicella, 1993; Pettit & Judd, 1992; Rice et al., 1986), and actively suppress (Liu et al., 2014; Liu & Russell, 2011), the immune response. Strain-specific antibodies are detected in serum, urethral exudates (Kearns et al., 1973; Tramont, 1977) and vaginal/cervical secretions (Hedges et al., 1999; O’Reilly et al., 1976; Tramont et al., 1980) of infected males and females. However, antibody production is low, it is not protective, and, with few exceptions, it is not significantly different among infected versus uninfected individuals (Hedges et al., 1999). The finding that antibodies directed against the gonococcal antigen RmpM (reduction modifiable protein M) block the bactericidal activity of other antibodies directed against N. gonorrhoeae (Rice et al., 1986), may explain, to a certain extent, the lack of protection seen. The presence and duration of antibody are also highly variable among individuals as well as between males and females (Hedges et al., 1999; Welch & O’Reilly, 1973).

Studies of the innate immune response during gonococcal infection provide additional clues as to how the gonococcus evades host defenses. Elegant studies demonstrate a role for complement inactivation and down regulation (Ngampasutadol et al., 2005, 2008a,b; Ram et al., 2001) in mediating infection (Edwards & Apicella, 2002; Jarvis, 1994; McQuillen et al., 1999). Gonococcal membrane-associated antimicrobial peptide efflux pumps (e.g. the Mtr protein complex) (Warner et al., 2007, 2008) are also shown to promote gonococcal survival during infection. However, the role of human phagocytic cells [neutrophils (PMNs) and macrophages] in gonococcal pathobiology is less well understood. The human neutrophil is clearly ineffective in resolving gonococcal infection, albeit evidence of stimulation of a respiratory burst, as well as a robust and prolonged response during gonococcal urethritis, exists (Apicella et al., 1996; Criss & Seifert, 2012; Seib et al., 2005; Simons et al., 2005). Infection studies performed in vitro using human PMNs indicate that there is an initial oxidative burst within the PMN upon infection resulting in death of 30–40% of the population (Simons et al., 2005). This is followed by replication of the resistant gonococci within the PMN and is associated with induction of anti-apoptosis, which prevents PMN death until 21–24 h after infection (Simons et al., 2006). At that time, the infected PMN is little more than a sac containing large numbers of gonococci (Simons et al., 2005). Understanding how the gonococcus evades PMN killing may lead to novel vaccine and therapeutic approaches. For example, data indicate that gonococcal surface proteins; porin (Bjerknes et al., 1995; Dehio et al., 2000; Haines et al., 1988, 1991; Lorenzen et al., 2000; Mosleh et al., 1998), pili (Stohl et al., 2013) and Opa (Ball & Criss, 2013); may have a substantive impact on the PMN response to infection. There are also data to indicate that a secreted gonococcal nuclease (Nuc) is involved in gonococcal survival by promoting escape from PMN nets (Juneau et al., 2015). Although the utility of porin, pili and Opa proteins as vaccine candidates is problematic (Jerse et al., 1994; Seifert et al., 1994; Swanson et al., 1987, 1988), targeting these (or other surface constituents) as an approach to treat gonococcal disease has not been examined. Similarly, the potential of Nuc as a vaccine antigen or a therapeutic target has not been examined. The ongoing development of multi-organ culture systems (outlined below) may provide novel ways to evaluate natural, and vaccine-induced, immune responses that involve phagocytic cells.

It is not clear if a single vaccine preparation, or elicitation of a single type of immune response, will be effective in both men and women (Edwards & Apicella, 2004). However, it is apparent that novel vaccination approaches that target and optimize specific aspects of the human response(s) to infection, in combination with scrutinous antigen selection, will be needed and may enable vaccine(s) development, provided a physiologically relevant model(s) of human disease and/or correlates of protection are established.

Models of infection and vaccine evaluation

Neisseria gonorrhoeae is an obligate human pathogen; therefore, humans serve as the only known natural reservoir for gonococcal infections. The in vivo human environment is difficult, if not impossible, to recapitulate in the laboratory, particularly given our incomplete understanding of the pathobiology of N. gonorrhoeae and the human response(s) to infection. Nevertheless, several infection models have been developed to investigate certain aspects of gonococcal infection, colonization and disease pathology (Arko, 1989; Edwards et al., 2000; Harvey et al., 1997; Jerse, 1999; Morales et al., 2006; Simons et al., 2005; Stephens, 1989; Stohl et al., 2005; Timmerman et al., 2005) (Table 2).

Table 2. Model systems for investigation of gonococcal infections and vaccine development.

Model Key features Pros Cons Key information to be gained
Human male challenge • Urethral infection of male volunteers • Mimics natural infection • Reflects disease in males • Evaluate gonococcal antigens that may only be highly expressed (in men) in vivo • May not reflect disease in females • Cost • Group size • Infection is terminated at the first signs of disease • Inability to investigate long term infections • Immunogenicity of antigens • Protective efficacy • Immune response(s) to infection • Role of specific gonococcal constituents in promoting infection/disease
Non-human primates • Infection of, and transmission between, males and females • Less subject to gonococcal host restrictions than are other laboratory animals • Ethical considerations • Limited availability • High costs • Immunogenicity of antigens • Protective efficacy
E2-mice • Administration of 17β-estradiol (E2) and antibiotics to female mice • Gonococcal infection of genital tract, and recovery of gonococci from infected animals for approximately 5–12 days • Abundant reagents available • More closely aligned with infection of males • Structural differences between mouse and female reproductive tract • Differences in the development, the activation, and the response to challenge between the murine and human immune systems • Less closely aligned with females • Absence of select human constituents as well as human-specific features of select human-murine orthologs • Immunogenicity of antigens • Compare adjuvants and routes of immunization
Guinea pigs and rabbits • Subcutaneous chambers implanted in animal • Reported to be better than mice when measuring immunogenicity of gonococcal antigens • Not been used in recent years • Immunogenicity of antigens • Compare adjuvants and routes of immunization
Primary Organ culture • E.g. fallopian tube, endometrium • Closely reflect natural sites of infection • Interactions between several host cell types can be evaluated • Typically does not allow the simultaneous analysis of innate and adaptive immune responses • Limited availability of human specimens • Technical expertise required to reproducibly procure these models • Cannot fully assess the contiguous variation in environmental factors occurring in vivo throughout the course of infection/disease • Ability of antibodies to inhibit antigen function (e.g. adherence, an intracellular survival, nutrient uptake)
Primary cell culture • E.g. cervical, endometrial, fallopian tube or male urethral epithelial cells, immune cells • Closely reflect natural sites of infection • Interactions between several host cell types can be evaluated • Typically does not allow the simultaneous analysis of innate and adaptive immune responses • Limited availability of human specimens • Technical expertise required to reproducibly procure these models • Cannot fully assess the contiguous variation in environmental factors occurring in vivo throughout the course of infection/disease • Ability of antibodies to inhibit antigen function • Ability to assess local production of innate immune effectors (cytokine responses, complement, antimicrobial peptides, etc.)
Immortalized/  cancel cell lines • E.g. cervical, endometrial or male urethral epithelial cells, immune cells • Relatively low cost • Readily available • Easily maintained • Interactions between several host cell types can be evaluated • Readily adaptable to high-throughput analyses • Typically do not allow the simultaneous analysis of innate and adaptive immune responses • Altered protein expression profiles relative to primary cells • Altered responses generated upon infection relative to the in vivo condition • Cannot fully assess the contiguous variation in environmental factors occurring in vivo throughout the course of infection/disease • Ability of antibodies to inhibit antigen function
Human/patient  fluids and tissues • Proteomic, genomic, metabolomic analyses of human specimens • Can be non-invasive, human-specific models • Limited availability of human specimens • Data obtained are restricted to a specific “snap shot” of infection/disease related to when the specimen is obtained • Identify human factors generated during infection and disease • Identify critical virulence determinants involved in immune induction or its suppression
“Human-on-a-chip” • Microfluidic cell culture chips that mimic multiple organs in the body • Non-invasive, human-specific models • Cost • Untested for gonorrhea, standardization likely will be required • System complexity • Human response(s) generated to infecting organisms
Immunotherapy • Treatment of infection using monoclonal antibodies or immune serum • Feasibility shown for delivery and targeting of antibodies to the genital tract • Targets available that are known to block colonization and essential metabolic activities of the bacteria • Cost • Untested for gonorrhea • Define the target, type, and level of antibody response needed to protect against, or reduce, mucosal infection or sequelae

The best system in which to study gonococcal pathogenesis is, of course, infected humans. In this regard, a human challenge model exists and involves experimental urethral infection of male volunteers. This model has been used to evaluate the protective role of vaccine candidates, to investigate the immune response(s) to infection and to examine the role of specific gonococcal constituents in promoting infection/disease (Cohen & Cannon, 1999; Cohen et al., 1994; Hobbs et al., 2011, 2013; Ramsey et al., 1994; Schmidt et al., 2001). This is also the optimal (in terms of mimicking natural infection) system available to test vaccine efficacy as well as to evaluate gonococcal antigens that only may be highly expressed (in men) in vivo. However, as with any scientific model, there are certain constraints that limit the utility of this infection model, including: (1) high associated costs, (2) a restriction to small group sizes and (3) infection is terminated at the first signs of disease and, thus, can only assess early stages of infection. Also of importance is that data obtained from infection in men likely does not translate to infection of women, as some pathways of infection are sex-specific (Edwards & Apicella, 2004). A similar model for investigations of gonococcal disease in women is ethically prohibited because of the potential for complications resulting from infection in females.

Several cell and organ culture systems have been used to study N. gonorrhoeae infection and offer an opportunity to define the potential of vaccine-induced antibodies to inhibit the function of the target protein (i.e. function inhibiting antibodies), such as by inhibiting adherence, invasion, colonization and/or biofilm formation. Primary human organ [e.g. fallopian tube (McGee et al., 1981), endometrium (Timmerman et al., 2005)] and epithelial cell [e.g. cervical (Edwards et al., 2001), endometrial (Timmerman et al., 2005), fallopian tube (Morales et al., 2006) or male urethral (Harvey et al., 1997)] cultures closely reflect natural sites of infection. However, it is impossible to simulate the continuum of numerous environmental changes (e.g. variable oxygen tensions, hormone levels, levels and presence of select proteins and/or cell types) that occur during in vivo infection in cell culture models. Moreover, the inability to simultaneously model aspects of the innate and the adaptive immune response in these systems has been a major drawback in vaccine research. These drawbacks can potentially be overcome in 3D cell culture systems which are being developed (Choi et al., 2015). The limited availability of human specimens and the technical expertise required to reproducibly procure these models has, however, resulted in the predominant use of immortalized or malignant human epithelial cell lines to study gonococcal–host interactions. Although cell lines are readily available and easily maintained, data obtained from the use of such cells may not be reflective of the epithelia encountered by the gonococcus in vivo. Continued laboratory maintenance, as well as cellular transformation, can alter the protein expression profiles of these cells as well as the responses generated upon infection [e.g. the absence of CR3 expression by immortal cervical and endometrial cell lines (Edwards et al., 2001)], and the altered production of pro-inflammatory mediators in immortalized human cervical and vaginal epithelial cells in response to gonococcal infection (Fichorova et al., 2001; Hedges et al., 1998).

It is generally believed that the limited host range exhibited by N. gonorrhoeae results largely from the specificity of an array of gonococcal proteins for human-specific targets that include: receptors for colonization [e.g. CR3 (Edwards et al., 2002); various CEACAMs (Sadarangani et al., 2011; Virji et al., 1996)]; iron-transport proteins [e.g. transferrin and lactoferrin (Lee & Schryvers, 1988)] and components of the immune system [e.g. C4BP and factor H (fH) (Ngampasutadol et al., 2008a,b)]. In this regard, our closest living relatives are the great apes (chimpanzees, orangutans and gorillas). Although, these non-human primates are less subject to gonococcal–host restrictions than are other laboratory animals, these models are rarely used because of ethical considerations, limited availability and high costs. Therefore, in the absence of a laboratory animal that is more closely related to humans, and with the abundance of reagents currently available to allow scientific analyses in mice, the use of a murine model to study gonococcal pathogenesis has gained increasing popularity. This model relies on the administration of 17β-estradiol (E2) and antibiotics to female mice, which enables gonococcal infection of the genital tract and recovery of gonococci from infected animals for approximately 5–12 days (reviewed in Jerse et al., 2011). The development of the estradiol-treated mouse infection model (here after referred to as the E2-mouse model) has provided an opportunity to test the immunogenicity of gonococcal antigens as well as to differentially compare adjuvants and routes of immunization.

The E2-mouse model of N. gonorrhoeae infection mirrors some aspects of human disease; however, it does not recapitulate infection of the human female reproductive tract or the immunological responses generated by infected women. Indeed, in many ways, data obtained from the use of this model are more closely aligned with infection of males than they are with females. For example, data obtained from experimental infections indicate that the pro-inflammatory cytokines interleukin (IL)-1β, IL-6, IL-8 and tumor necrosis factor (TNF)-α are elevated in infected males when compared to uninfected control volunteers (Ramsey et al., 1995). Similar data are obtained upon analyses of urethral exudates from men with culture-documented gonorrhea, from cell culture supernatants collected from gonococci-infected primary (human) male urethral epithelial cells (Harvey et al., 2002), and from genital secretions of E2-treated female mice (Packiam et al., 2010). Conversely, analyses of vaginal and cervical fluids collected from women with culture-documented gonococcal cervicitis reveal that local levels of these inflammatory mediators are not elevated during natural infection (Hedges et al., 1998). Neisseria gonorrhoeae infection of primary (Edwards & Apicella, 2004), but not immortalized (Fichorova et al., 2001), human cervical epithelial cells yields results comparable to those observed in naturally-infected females (Hedges et al., 1998). More recently, Russell and co-workers have shown that infection of mice, or mouse vaginal explants, results in increased levels of IL-17A, IL-23, IL-6 and TNF-α, whereas IL-12 (associated with the development of a Th1-mediated response) was not detected (Feinen et al., 2010). Based on these data, this group has proposed that (predominately αβ) T-cells of the Th17 lineage play a central role in the immune response to N. gonorrhoeae, resulting in the production of proinflammatory cytokines and chemokines, and the recruitment of neutrophils and other innate defense factors to the site of infection. Consistent with these data, Gagliardi et al. (2011) report increased serum concentrations of IL-17A and IL-23 (as well as IFN-γ) in infected humans; however, it is important to note that of the 27 patients with gonorrhea examined, 26 of these were men, as were all of the control (uninfected) subjects. To our knowledge, the levels of Th17-related cytokines have not been measured in N. gonorrhoeae-infected women. However, IL-1β, IL-6 and TGF-β (associated with Th17 induction) are not elevated in vaginal and cervical fluids of infected women (Hedges et al., 1998). Thereby, these data suggest that, unlike infection in mice, Th17 cells likely may not play a substantive role in the immune response to gonococcal infection in women, albeit Th17 cell responses do appear to play a role in the immune response to infection in men.

Important structural/functional differences exist between the female reproductive tract of women and that of mice (Rendi et al., 2012). Such differences also may, in part, contribute to difficulties in translating data obtained from the E2-mouse model of infection to females. Of note, is the presence of the squamocolumnar junction/transformation zone (i.e. the distinctly abrupt area of transition from the columnar, to the stratified squamous, epithelia of the endo- and ectocervix, respectively) in humans and its absence in mice. Although the epithelia of the female reproductive tract lack mucosa-associated lymphoreticular tissue (i.e. MALT), organized areas of lymphoid aggregates (comprised of Langerhans, plasma, T- and B-cells) are concentrated within the transformation zone as well as within the cervix and endometrium (Edwards & Morris, 1985; Pudney et al., 2005; Yeaman et al., 2001). Therefore, it is suggested that the transformation zone exists as a unique immunologically active site that likely plays an important role in antigen uptake, lymphocyte recruitment and differentiation, as well as in prohibiting ascension of potential pathogens to the uterine cavity. Similar studies performed in mice indicate that “lymphoid nodules” and B-cells are not present within the epithelia of the murine female reproductive tract (Parr & Parr, 1991). Further investigations should be performed to verify these data, as this may have important implications with regard to the utility of the mouse model for antigen screening and pre-clinical vaccine studies, if women are to be the targeted population. For example, it is suggested that vaccines intended to induce protective immunity within the female reproductive tract should be targeted to the transformation zone/ectocervix for a cytotoxic T-cell-mediated response or to the endocervix for a local antibody-mediated response, neither of which may be possible to evaluate in a mouse model (Pudney et al., 2005). It is also important to note that there are substantive differences in the development, the activation and the response to challenge between the human and murine immune systems (Mestas & Hughes, 2004). Whereas mice have orthologs of many immunologically important human molecules, similarity in function does not always relate to causality. That is, immunological function/outcomes can be achieved in different species by divergent routes, and the potential protective effects of vaccines against targets that interact with host-restricted molecules might be under- or over-estimated in normal mice.

Looking forward

Pre-clinical development and evaluation of vaccines are typically based on protection (e.g. decreases in bacterial load or in illness/symptoms) in animal models, or by using correlates/surrogates of protection (i.e. measureable evidence of protection from infection or disease, e.g. altered mucosal and cellular immune responses). This has led to an over-reliance on the use of animals, in particular mice, by which to study human (-specific) diseases. This is despite the fact that most trials in animals do not show a similar outcome in human clinical trials (Hackam & Redelmeier, 2006; Knight, 2007; Pound et al., 2004), a problem that is particularly true for those trials examining inflammation (Seok et al., 2013). This can be attributed, at least in part, to the inability of some animal models to accurately replicate the human disease that the animal is intended to model. Given that correlates of protection for N. gonorrhoeae are unknown, and there is no animal model that accurately mimics the complexity of human disease in males and females, this begs the question, where do we go from here when there are valid concerns that a time of untreatable gonorrhea may be quickly approaching?

In terms of models systems for vaccine development and pre-clinical trials for exclusive human pathogens, such as the gonococcus, it is important that the model is selected, and data are interpreted, with specific questions and intended outcomes in mind [i.e. relating to immunogenicity, toxicity or protection, and the group (male and/or female) being targeted]. Therefore, data obtained from the use of laboratory models should mimic as closely as possible and, thereby, complement data obtained from human sources. Together, this will ensure that the data obtained from such models are relevant and can be translated to the human male and/or female as effectively as possible. For example, the use of IL-12-encapsulated microspheres has been proposed as a therapeutic option for the treatment of gonorrhea based on data obtained from studies performed in mice. Among the functions ascribed to IL-12 is its ability to antagonize some of the effects of IL-10 and TGF-β (Liu et al., 2013). In that IL-10 and TGF-β are shown to be already low in women with gonococcal disease (Hedges et al., 1999), it seems prudent to question whether such a treatment might exacerbate infection in women, rather than aid in its resolution. Whereas IL-12 therapy may, in theory, provide one solution to treat gonorrhea in males, in practice it may not be well accepted because of patient discomfort and, thereby, patient compliance.

The E2-mouse model permits investigations in a mammalian host and provides a valuable way to test antigen immunogenicity, using different adjuvants and routes of immunization. Subcutaneous chambers implanted in Guinea pigs and rabbits have also been used for such studies and are reported to be comparatively better than mice when measuring immunogenicity of gonococcal antigens (Arko, 1972). Although these models have not been used in recent years, they could be re-evaluated for their utility in determining correlates of protection, the potential immunogenicity of select gonococcal antigens and/or to conduct preclinical toxicity studies for new therapeutic regimens to treat gonococcal disease. The subcutaneous Guinea pig infection model, as described, does not require the administration of E2 and antibiotics to establish infection (Arko, 1972). Therefore, the comparative use of these models could provide insights into any extraneous effects resulting from E2 (or antibiotic) administration to mice (e.g. immune suppression), which have the potential to skew data interpretation and, in turn, the ability to translate such data to N. gonorrhoeae vaccine development for use in humans.

Given that currently used mammalian models (the E2-mouse model and male human volunteers) may not be physiologically relevant models of female infection, it may also be important to take the focus off targeting human females for vaccination. Recently, the potential impact of “hypothetical” vaccines was modeled (Craig et al., 2015). This study indicated that the sex of those vaccinated was unimportant provided the same percentage of the eligible population is vaccinated. That is, a similar reduction in gonorrhea prevalence was predicted for potential vaccines (with 50% or 100% efficacy and 20 years duration of protection) whether 50% of all 13-year-olds, or 100% of male or female 13-year-olds, were vaccinated (Craig et al., 2015).

In light of the anatomical and physiological differences that exist between humans and mice (the only animal model presently used), there is a need for the increased use of humanized models, albeit even humanized animal models of infection may not be sufficient to overcome the numerous obstacles associated with gonococcal vaccine development and will not compensate for anatomical differences among species. Given the propensity of the gonococcus to subvert human-specific molecules to promote infection and disease, data obtained from human sources could, and should, be used to guide the development of such animal models. Total proteomic, genomic, metabolomic analyses of human specimens (e.g. patient fluids and tissues) offer a means of identifying human factors contributing to infection but also the human response(s) generated to infecting organisms. Similar analyses of bacteria isolated from these patients would also identify critical virulence determinants promoting bacterial survival/proliferation and/or that play a role in immune induction or its suppression. The increased speed and lowered costs now associated with these “big data” techniques presently make these approaches more accessible to investigators and, thereby, increase their feasible contribution to vaccine development. Moreover, the relevance of knowledge gained through the ex vivo analysis of human subjects cannot be under stated. Focused quantitative analyses to verify/support data obtained by ex vivo analyses of human subjects can be achieved by using primary human cell and organ cultures as well as animal models. Moreover, multi-organ cultures (MOCs) could be adapted to incorporate the combined use of primary human epithelial and immune cells (or tissues) into a single experimental model. MOC models are currently being used in pre-clinical drug toxicity studies and offer an excellent opportunity to study the human immune responses generated during infection. The development of MOCs and “organs-/humans-on-chips” also provide alternative non-invasive, human-specific models by which data obtained from patient specimens can be further evaluated (Bhatia & Ingber, 2014).

An increased focus on human samples and humanized animal models may provide insights into what is required for protective immunity against gonorrhea and help define a correlate of protection. Evaluating immunotherapy may enable us to begin to define the target, type and level of antibody response needed to protect against, or reduce, mucosal infection or sequelae. Several studies support the feasibility of immunotherapy to treat sexually transmitted infections, in terms of effective delivery and targeting of antibodies to the genital tract, leading to clearance of infection and/or prevention of disease (reviewed in Naz, 2012). For example, mice given prophylactic or therapeutic intravenous hu2c monoclonal antibodies (mAbs) were completely protected from infection-related mortality after vaginal challenge with a lethal dose of HSV-1. Almost complete protection was also seen when hu2c was given intraperitoneally after infection with a multidrug-resistant HSV-1 patient isolate (Krawczyk et al., 2013). Rhesus macaques given neutralizing mAb intravenously were protected against infection or disease manifestations after vaginal challenge with a pathogenic HIV-1/SIV chimeric virus (Mascola et al., 2000). In humans, combined immunochemotherapy with the chimeric human/mouse mAb rituximab has been used to successfully treat primary non-Hodgkin's lymphoma of the uterus, cervix and parametrium (Su et al., 2008), and the humanized mAb Trastuzumab provided relief of symptoms, and prolonged survival, in patients with endometrial carcinoma (Santin et al., 2008). Various delivery systems (e.g. liposomes, microspheres, nanoparticles, nanogels, bionanocapsules) are under investigation (Buss et al., 2012) that may enable monoclonal antibodies to be directly delivered to the genital tract in the future. Potential gonococcal antigen targets that can be exploited for antibody therapy also exist. For example, antibodies to gonococcal phospholipase D (PLD), a key regulator of infection of human cervical epithelia (Apicella & Edwards, 2007), are able to decrease adherence to and invasion of primary cervical cells in vitro (Edwards et al., 2003). Antibodies against a truncated, non-glycosylated, recombinant form of the AniA protein are able of block its nitrite reductase function in a whole cell assay (Shewell et al., 2013). Several other examples are listed in Table 1, and a combination of antibodies that are able to block colonization and essential metabolic activities of the bacteria may have a significant impact in preventing infection, transmission and/or disease. Even a modest reduction in infection or sequelae by immunotherapy may provide a significant impact on prevalence, similar to what has been reported for “hypothetical” vaccines, in which even a vaccine with modest efficacy could have a substantial impact on gonorrhea prevalence (Craig et al., 2015). Although monoclonal antibody therapy is expensive and likely to be of limited utility for the population that needs it most, it may aid in understanding required correlates of protection and may serve as a last line of therapy for untreatable N. gonorrhoeae strains.

Conclusions

To prevent the increased prevalence of gonorrhea, and its associated disease sequelae, there is an urgent need for increased investment in antibiotic and vaccine development. Future research should also focus on the development of new models of infection and the use of all available tools without an over-reliance on one particular model or method. No infection model is without its limitations, but it may be possible to overcome many of the current limitations by using a combined approach and by placing an emphasis on collecting and incorporating human (or human source)-derived data. Novel approaches may also be required, such as the use of “organs-/humans-on-chips” or the evaluation of immunotherapy, to help address the many unanswered questions regarding the mechanisms of protective immunity and what is required to combat the human specific pathogen N. gonorrhoeae. In the absence of a correlate of protection, it is difficult to predict the success of candidate gonococcal antigens based on pre-clinical data. Therefore, it may be that human clinical trials are needed, using the best available antigens (based on immunogenicity and antibody function), to provide evidence of effectiveness and provide a pathway to vaccine licensure. A similar approach was used during the development of the acellular pertussis vaccine, for which the protective mechanism has not been fully elucidated (Farizo et al., 2014). Given the high cost of this approach, it is likely that public–private partnerships will be required to drive development of a gonococcal vaccine. In this regard, the recent emergence of untreatable N. gonorrhoeae strains may fuel collaborative efforts towards vaccine development. An increase in the number of untreatable gonorrhea cases may, in the future, also force changes in the way gonococcal vaccines are brought to the public; similar to the way the outbreaks of N. meningitidis infections at Princeton University and the University of California, Santa Barbara resulted in FDA approval of the meningococcal vaccine Bexsero to at-risk populations at these universities (only) (McNamara et al., 2015; Seib et al., 2015).

Acknowledgements

JLE and KLS contributed equally to this paper and are co-senior authors.

Declaration of interest

This work was supported by the Australian National Health and Medical Research Council (NHMRC) [Project Grant 1028326 and Career Development Fellowship to KLS; Program Grant 565526 to MPJ]; and the National Institutes of Health (USA) [NIAID Grants 1R01AI076398 to JLE; and R01AI108255 and R01AI09320 to MAA]. The authors report no conflicts of interest. The authors alone are responsible for the content and writing of this article.

References

  1. Adamiak P, Beddek AJ, Pajon R, Schryvers AB. Patterns of sequence variation within the Neisseria meningitidis lactoferrin binding proteins. Biochem Cell Biol. 2012;90:339–50. doi: 10.1139/o11-076. [DOI] [PubMed] [Google Scholar]
  2. Aledort JE, Hook EW, III, Weinstein MC, Goldie SJ. The cost effectiveness of gonorrhea screening in urban emergency departments. Sex Transm Dis. 2005;32:425–36. doi: 10.1097/01.olq.0000154501.22566.fa. [DOI] [PubMed] [Google Scholar]
  3. Apicella MA, Edwards JL. 2007. Vaccine and compositions for the prevention and treatment of neisserial infections (Patent US7261901) [Google Scholar]
  4. Apicella MA, Ketterer M, Lee FK, et al. The pathogenesis of gonococcal urethritis in men: confocal and immunoelectron microscopic analysis of urethral exudates from men infected with Neisseria gonorrhoeae . J Infect Dis. 1996;173:636–46. doi: 10.1093/infdis/173.3.636. [DOI] [PubMed] [Google Scholar]
  5. Arko RJ. Neisseria gonorrhoeae: experimental infection of laboratory animals. Science. 1972;177:1200–1. doi: 10.1126/science.177.4055.1200. [DOI] [PubMed] [Google Scholar]
  6. Arko RJ. Animal models for pathogenic Neisseria species. Clin Microbiol Rev. 1989;2:S56–9. doi: 10.1128/cmr.2.suppl.s56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Ball LM, Criss AK. Constitutively Opa-expressing and Opa-deficient Neisseria gonorrhoeae strains differentially stimulate and survive exposure to human neutrophils. J Bacteriol. 2013;195:2982–90. doi: 10.1128/JB.00171-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Barry PM, Klausner JD. The use of cephalosporins for gonorrhea: the impending problem of resistance. Expert Opin Pharmacother. 2009;10:555–77. doi: 10.1517/14656560902731993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nat Biotechnol. 2014;32:760–72. doi: 10.1038/nbt.2989. [DOI] [PubMed] [Google Scholar]
  10. Biswas GD, Anderson JE, Chen CJ, et al. Identification and functional characterization of the Neisseria gonorrhoeae lbpB gene product. Infect Immun. 1999;67:455–9. doi: 10.1128/iai.67.1.455-459.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bjerknes R, Guttormsen HK, Solberg CO, Wetzler LM. Neisserial porins inhibit human neutrophil actin polymerization, degranulation, opsonin receptor expression, and phagocytosis but prime the neutrophils to increase their oxidative burst. Infect Immun. 1995;63:160–7. doi: 10.1128/iai.63.1.160-167.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bolan GA, Sparling PF, Wasserheit JN. The emerging threat of untreatable gonococcal infection. N Engl J Med. 2012;366:485–7. doi: 10.1056/NEJMp1112456. [DOI] [PubMed] [Google Scholar]
  13. Boslego JW, Tramont EC, Chung RC, et al. Efficacy trial of a parenteral gonococcal pilus vaccine in men. Vaccine. 1991;9:154–62. doi: 10.1016/0264-410x(91)90147-x. [DOI] [PubMed] [Google Scholar]
  14. Buss NA, Henderson SJ, McFarlane M, et al. Monoclonal antibody therapeutics: history and future. Curr Opin Pharmacol. 2012;12:615–22. doi: 10.1016/j.coph.2012.08.001. [DOI] [PubMed] [Google Scholar]
  15. Callaghan MJ, Lewis S, Sadarangani M, et al. Potential of recombinant opa proteins as vaccine candidates against hyperinvasive meningococci. Infect Immun. 2011;79:2810–18. doi: 10.1128/IAI.01338-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. CDC. (2013). Antibiotic resistance threats in the United States, 2013. [On-line]. Available from: http://www.cdc.gov/drugresistance/threat-report-2013/pdf/ar-threats-2013-508.pdf [last accessed 7 Mar 2014]
  17. CDC Update to CDC's Sexually transmitted diseases treatment guidelines, 2010: oral cephalosporins no longer a recommended treatment for gonococcal infections. 2012 [PubMed] [Google Scholar]
  18. CDC. (2014). Sexually transmitted diseases surveillance 2013. [On-line]. Available from: http://www.cdc.gov/std/stats13/default.htm [last accessed 7 Mar 2014]
  19. Chen A, Boulton IC, Pongoski J, et al. Induction of HIV-1 long terminal repeat-mediated transcription by Neisseria gonorrhoeae . AIDS. 2003;17:625–8. doi: 10.1097/00002030-200303070-00019. [DOI] [PubMed] [Google Scholar]
  20. Choi J, Lee EK, Choo J, et al. Micro 3D cell culture systems for cellular behavior studies: culture matrices, devices, substrates, and in-situ sensing methods. Biotechnol J. 2015 doi: 10.1002/biot.201500092. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  21. Clark VL, Knapp JS, Thompson S, Klimpel KW. Presence of antibodies to the major anaerobically induced gonococcal outer membrane protein in sera from patients with gonococcal infections. Microb Pathog. 1988;5:381–90. doi: 10.1016/0882-4010(88)90038-1. [DOI] [PubMed] [Google Scholar]
  22. Cohen MS, Cannon JG, Jerse AE, et al. Human experimentation with Neisseria gonorrhoeae: rationale, methods, and implications for the biology of infection and vaccine development. J Infect Dis. 1994;169:532–7. doi: 10.1093/infdis/169.3.532. [DOI] [PubMed] [Google Scholar]
  23. Cohen MS, Cannon JG. Human experimentation with Neisseria gonorrhoeae: progress and goals. J Infect Dis. 1999;179:S375–9. doi: 10.1086/513847. [DOI] [PubMed] [Google Scholar]
  24. Cohen MS, Hoffman IF, Royce RA, et al. Reduction of concentration of HIV-1 in semen after treatment of urethritis: implications for prevention of sexual transmission of HIV-1. AIDSCAP Malawi Research Group. Lancet. 1997;349:1868–73. doi: 10.1016/s0140-6736(97)02190-9. [DOI] [PubMed] [Google Scholar]
  25. Cole JG, Jerse AE. Functional characterization of antibodies against Neisseria gonorrhoeae opacity protein loops. PLoS One. 2009;4:e8108. doi: 10.1371/journal.pone.0008108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cornelissen CN, Hollander A. TonB-dependent transporters expressed by Neisseria gonorrhoeae . Front Microbiol. 2011;2:117. doi: 10.3389/fmicb.2011.00117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Craig AP, Gray RT, Edwards JL, et al. The potential impact of vaccination on the prevalence of gonorrhea. Vaccine. 2015;33:4520–5. doi: 10.1016/j.vaccine.2015.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Criss AK, Seifert HS. A bacterial siren song: intimate interactions between Neisseria and neutrophils. Nat Rev Microbiol. 2012;10:178–90. doi: 10.1038/nrmicro2713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. de Jonge MI, Hamstra HJ, Jiskoot W, et al. Intranasal immunisation of mice with liposomes containing recombinant meningococcal OpaB and OpaJ proteins. Vaccine. 2004;22:4021–8. doi: 10.1016/j.vaccine.2004.03.047. [DOI] [PubMed] [Google Scholar]
  30. Dehio C, Gray-Owen SD, Meyer TF. Host cell invasion by pathogenic Neisseriae. Subcell Biochem. 2000;33:61–96. doi: 10.1007/978-1-4757-4580-1_4. [DOI] [PubMed] [Google Scholar]
  31. Edwards JL, Apicella MA. The role of lipooligosaccharide in Neisseria gonorrhoeae pathogenesis of cervical epithelia: lipid A serves as a C3 acceptor molecule. Cell Microbiol. 2002;4:585–98. doi: 10.1046/j.1462-5822.2002.00212.x. [DOI] [PubMed] [Google Scholar]
  32. Edwards JL, Apicella MA. The molecular mechanisms used by Neisseria gonorrhoeae to initiate infection differ between men and women. Clin Microbiol Rev. 2004;17:965–81. doi: 10.1128/CMR.17.4.965-981.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Edwards JL, Apicella MA. Neisseria gonorrhoeae PLD directly interacts with Akt kinase upon infection of primary, human, cervical epithelial cells. Cell Microbiol. 2006;8:1253–71. doi: 10.1111/j.1462-5822.2006.00707.x. [DOI] [PubMed] [Google Scholar]
  34. Edwards JL, Brown EJ, Ault KA, Apicella MA. The role of complement receptor 3 (CR3) in Neisseria gonorrhoeae infection of human cervical epithelia. Cell Microbiol. 2001;3:611–22. doi: 10.1046/j.1462-5822.2001.00140.x. [DOI] [PubMed] [Google Scholar]
  35. Edwards JL, Brown EJ, Uk-Nham S, et al. A co-operative interaction between Neisseria gonorrhoeae and complement receptor 3 mediates infection of primary cervical epithelial cells. Cell Microbiol. 2002;4:571–84. doi: 10.1046/j.1462-5822.2002.t01-1-00215.x. [DOI] [PubMed] [Google Scholar]
  36. Edwards JL, Entz DD, Apicella MA. Gonococcal phospholipase D modulates the expression and function of complement receptor 3 in primary cervical epithelial cells. Infect Immun. 2003;71:6381–91. doi: 10.1128/IAI.71.11.6381-6391.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Edwards JL, Shao JQ, Ault KA, Apicella MA. Neisseria gonorrhoeae elicits membrane ruffling and cytoskeletal rearrangements upon infection of primary human endocervical and ectocervical cells. Infect Immun. 2000;68:5354–63. doi: 10.1128/iai.68.9.5354-5363.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Edwards JN, Morris HB. Langerhans' cells and lymphocyte subsets in the female genital tract. Br J Obstet Gynaecol. 1985;92:974–82. doi: 10.1111/j.1471-0528.1985.tb03080.x. [DOI] [PubMed] [Google Scholar]
  39. Eyre JWH, Stewart BH. The treatment of gonococcus infections by vaccines. Lancet. 1909;174:76–81. [Google Scholar]
  40. Falsetta ML, Bair TB, Ku SC, et al. Transcriptional profiling identifies the metabolic phenotype of gonococcal biofilms. Infect Immun. 2009;77:3522–32. doi: 10.1128/IAI.00036-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Farizo KM, Burns DL, Finn TM, et al. Clinical evaluation of pertussis vaccines: US Food and Drug Administration regulatory considerations. J Infect Dis. 2014;209:S28–31. doi: 10.1093/infdis/jit532. [DOI] [PubMed] [Google Scholar]
  42. Farley TA, Cohen DA, Elkins W. Asymptomatic sexually transmitted diseases: the case for screening. Prev Med. 2003;36:502–9. doi: 10.1016/s0091-7435(02)00058-0. [DOI] [PubMed] [Google Scholar]
  43. Feinen B, Jerse AE, Gaffen SL, Russell MW. Critical role of Th17 responses in a murine model of Neisseria gonorrhoeae genital infection. Mucosal Immunol. 2010;3:312–21. doi: 10.1038/mi.2009.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Fichorova RN, Desai PJ, Gibson FC, III, Genco CA. Distinct proinflammatory host responses to Neisseria gonorrhoeae infection in immortalized human cervical and vaginal epithelial cells. Infect Immun. 2001;69:5840–8. doi: 10.1128/IAI.69.9.5840-5848.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Fowler T, Caley M, Johal R, et al. Previous history of gonococcal infection as a risk factor in patients presenting with gonorrhoea. Int J STD AIDS. 2010;21:277–8. doi: 10.1258/ijsa.2009.009235. [DOI] [PubMed] [Google Scholar]
  46. Fox KK, Thomas JC, Weiner DH, et al. Longitudinal evaluation of serovar-specific immunity to Neisseria gonorrhoeae . Am J Epidemiol. 1999;149:353–8. doi: 10.1093/oxfordjournals.aje.a009820. [DOI] [PubMed] [Google Scholar]
  47. Gagliardi MC, Starnino S, Teloni R, et al. Circulating levels of interleukin-17A and interleukin-23 are increased in patients with gonococcal infection. FEMS Immunol Med Microbiol. 2011;61:129–32. doi: 10.1111/j.1574-695X.2010.00759.x. [DOI] [PubMed] [Google Scholar]
  48. Gallo MF, Macaluso M, Warner L, et al. Bacterial vaginosis, gonorrhea, and chlamydial infection among women attending a sexually transmitted disease clinic: a longitudinal analysis of possible causal links. Ann Epidemiol. 2012;22:213–20. doi: 10.1016/j.annepidem.2011.11.005. [DOI] [PubMed] [Google Scholar]
  49. Garvin LE, Begum AAB, Bash MC, Jerse AE. (2010). Elicit broadly cross reactive, bactericidal antibodies against Neisseria gonorrhoeae. (Abstract P235), abstr International Pathogenic Neisseria Conference, Banff, Canada. [On-line]. Available from: http://neisseria.org/ipnc/history.shtml [last accessed 7 Mar 2014]
  50. Greenberg L, Diena BB, Ashton FA, et al. Gonococcal vaccine studies in Inuvik. Can J Public Health. 1974;65:29–33. [PubMed] [Google Scholar]
  51. Gulati S, Agarwal S, Vasudhev S, et al. Properdin is critical for antibody-dependent bactericidal activity against Neisseria gonorrhoeae that recruit C4b-binding protein. J Immunol. 2012;188:3416–25. doi: 10.4049/jimmunol.1102746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Gulati S, McQuillen DP, Mandrell RE, et al. Immunogenicity of Neisseria gonorrhoeae lipooligosaccharide epitope 2C7, widely expressed in vivo with no immunochemical similarity to human glycosphingolipids. J Infect Dis. 1996;174:1223–37. doi: 10.1093/infdis/174.6.1223. [DOI] [PubMed] [Google Scholar]
  53. Gulati S, Zheng B, Reed GW, et al. Immunization against a saccharide epitope accelerates clearance of experimental gonococcal infection. PLoS Pathog. 2013;9:e1003559. doi: 10.1371/journal.ppat.1003559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Hackam DG, Redelmeier DA. Translation of research evidence from animals to humans. JAMA. 2006;296:1731–2. doi: 10.1001/jama.296.14.1731. [DOI] [PubMed] [Google Scholar]
  55. Haghi F, Peerayeh SN, Siadat SD, Zeighami H. Recombinant outer membrane secretin PilQ(406-770) as a vaccine candidate for serogroup B Neisseria meningitidis . Vaccine. 2012;30:1710–14. doi: 10.1016/j.vaccine.2011.12.076. [DOI] [PubMed] [Google Scholar]
  56. Haines KA, Reibman J, Tang XY, et al. Effects of protein I of Neisseria gonorrhoeae on neutrophil activation: generation of diacylglycerol from phosphatidylcholine via a specific phospholipase C is associated with exocytosis. J Cell Biol. 1991;114:433–42. doi: 10.1083/jcb.114.3.433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Haines KA, Yeh L, Blake MS, et al. Protein I, a translocatable ion channel from Neisseria gonorrhoeae, selectively inhibits exocytosis from human neutrophils without inhibiting O2-generation. J Biol Chem. 1988;263:945–51. [PubMed] [Google Scholar]
  58. Handsfield HH, Lipman TO, Harnisch JP, et al. Asymptomatic gonorrhea in men. Diagnosis, natural course, prevalence and significance. N Engl J Med. 1974;290:117–23. doi: 10.1056/NEJM197401172900301. [DOI] [PubMed] [Google Scholar]
  59. Harvey HA, Ketterer MR, Preston A, et al. Ultrastructural analysis of primary human urethral epithelial cell cultures infected with Neisseria gonorrhoeae . Infect Immun. 1997;65:2420–7. doi: 10.1128/iai.65.6.2420-2427.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Harvey HA, Post DM, Apicella MA. Immortalization of human urethral epithelial cells: a model for the study of the pathogenesis of and the inflammatory cytokine response to Neisseria gonorrhoeae infection. Infect Immun. 2002;70:5808–15. doi: 10.1128/IAI.70.10.5808-5815.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Hedges SR, Mayo MS, Mestecky J, et al. Limited local and systemic antibody responses to Neisseria gonorrhoeae during uncomplicated genital infections. Infect Immun. 1999;67:3937–46. doi: 10.1128/iai.67.8.3937-3946.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Hedges SR, Sibley DA, Mayo MS, et al. Cytokine and antibody responses in women infected with Neisseria gonorrhoeae: effects of concomitant infections. J Infect Dis. 1998;178:742–51. doi: 10.1086/515372. [DOI] [PubMed] [Google Scholar]
  63. Hobbs MM, Anderson JE, Balthazar JT, et al. Lipid A's structure mediates Neisseria gonorrhoeae fitness during experimental infection of mice and men. MBio. 2013;4:e00892–13. doi: 10.1128/mBio.00892-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Hobbs MM, Sparling PF, Cohen MS, et al. Experimental gonococcal infection in male volunteers: cumulative experience with Neisseria gonorrhoeae strains FA1090 and MS11mkC. Front Microbiol. 2011;2:123. doi: 10.3389/fmicb.2011.00123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Holst J, Oster P, Arnold R, et al. Vaccines against meningococcal serogroup B disease containing outer membrane vesicles (OMV): lessons from past programs and implications for the future. Hum Vaccin Immunother. 2013;9:1241–53. doi: 10.4161/hv.24129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Hook EW, Hansfield HH. Gonoccoal Infection in the Adult. In: Holmes KK, editor. Sexually transmitted diseases. New York: McGraw-Hill; 2008. pp. 627–45. [Google Scholar]
  67. Huppert JS, Goodman E, Khoury J, Slap G. Sexually transmitted infection testing and screening in hospital-based primary care visits by women. Obstet Gynecol. 2005;105:390–6. doi: 10.1097/01.AOG.0000151129.47746.be. [DOI] [PubMed] [Google Scholar]
  68. Jarvis GA. Analysis of C3 deposition and degradation on Neisseria meningitidis and Neisseria gonorrhoeae . Infect Immun. 1994;62:1755–60. doi: 10.1128/iai.62.5.1755-1760.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Jerse AE, Bash MC, Russell MW. Vaccines against gonorrhea: current status and future challenges. Vaccine. 2014;32:1579–87. doi: 10.1016/j.vaccine.2013.08.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Jerse AE, Cohen MS, Drown PM, et al. Multiple gonococcal opacity proteins are expressed during experimental urethral infection in the male. J Exp Med. 1994;179:911–20. doi: 10.1084/jem.179.3.911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Jerse AE, Deal CD. Vaccine research for gonococcal infections: where are we? Sex Transm Infect. 2013;89:iv63–8. doi: 10.1136/sextrans-2013-051225. [DOI] [PubMed] [Google Scholar]
  72. Jerse AE, Wu H, Packiam M, et al. Estradiol-treated female mice as surrogate hosts for Neisseria gonorrhoeae genital tract infections. Front Microbiol. 2011;2:107. doi: 10.3389/fmicb.2011.00107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Jerse AE. Experimental gonococcal genital tract infection and opacity protein expression in estradiol-treated mice. Infect Immun. 1999;67:5699–708. doi: 10.1128/iai.67.11.5699-5708.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Johnson LF, Alkema L, Dorrington RE. A Bayesian approach to uncertainty analysis of sexually transmitted infection models. Sex Transm Infect. 2010;86:169–74. doi: 10.1136/sti.2009.037341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Juneau RA, Stevens JS, Apicella MA, Criss AK. A thermonuclease of Neisseria gonorrhoeae enhances bacterial escape from killing by neutrophil extracellular traps. J Infect Dis. 2015;212:316–24. doi: 10.1093/infdis/jiv031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Kearns DH, O'Reilly RJ, Lee L, Welch BG. Secretory IgA antibodies in the urethral exudate of men with uncomplicated urethritis due to Neisseria gonorrhoeae . J Infect Dis. 1973;127:99–101. doi: 10.1093/infdis/127.1.99. [DOI] [PubMed] [Google Scholar]
  77. Kilian M, Reinholdt J. A hypothetical model for the development of invasive infection due to IgA1 protease-producing bacteria. Adv Exp Med Biol. 1987;216B:1261–9. [PubMed] [Google Scholar]
  78. Kirkcaldy RD, Weinstock HS, Moore PC, et al. The efficacy and safety of gentamicin plus azithromycin and gemifloxacin plus azithromycin as treatment of uncomplicated gonorrhea. Clin Infect Dis. 2014;59:1083–91. doi: 10.1093/cid/ciu521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Knight A. Systematic reviews of animal experiments demonstrate poor human clinical and toxicological utility. Altern Lab Anim. 2007;35:641–59. doi: 10.1177/026119290703500610. [DOI] [PubMed] [Google Scholar]
  80. Krawczyk A, Arndt MA, Grosse-Hovest L, et al. Overcoming drug-resistant herpes simplex virus (HSV) infection by a humanized antibody. Proc Natl Acad Sci USA. 2013;110:6760–5. doi: 10.1073/pnas.1220019110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Lahra MM, Ryder N, Whiley DM. A new multidrug-resistant strain of Neisseria gonorrhoeae in Australia. N Engl J Med. 2014;371:1850–1. doi: 10.1056/NEJMc1408109. [DOI] [PubMed] [Google Scholar]
  82. Lee BC, Schryvers AB. Specificity of the lactoferrin and transferrin receptors in Neisseria gonorrhoeae . Mol Microbiol. 1988;2:827–9. doi: 10.1111/j.1365-2958.1988.tb00095.x. [DOI] [PubMed] [Google Scholar]
  83. Levine WC, Pope V, Bhoomkar A, et al. Increase in endocervical CD4 lymphocytes among women with nonulcerative sexually transmitted diseases. J Infect Dis. 1998;177:167–74. doi: 10.1086/513820. [DOI] [PubMed] [Google Scholar]
  84. Lewis LA, Burrowes E, Rice PA, Ram S. Interactions of Neisseria with complement. In: Genco CA, Wetzler L, editors. Neisseria: molecular mechanisms of pathogenesis. Norfolk: Caister Academic Press; 2010. pp. 123–44. [Google Scholar]
  85. Lewis DA, Sriruttan C, Muller EE, et al. Phenotypic and genetic characterization of the first two cases of extended-spectrum-cephalosporin-resistant Neisseria gonorrhoeae infection in South Africa and association with cefixime treatment failure. J Antimicrob Chemother. 2013;68:1267–70. doi: 10.1093/jac/dkt034. [DOI] [PubMed] [Google Scholar]
  86. Li G, Jiao H, Jiang G, et al. Neisseria gonorrhoeae NspA induces specific bactericidal and opsonic antibodies in mice. Clin Vaccine Immunol. 2011;18:1817–22. doi: 10.1128/CVI.05245-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Liu Y, Egilmez NK, Russell MW. Enhancement of adaptive immunity to Neisseria gonorrhoeae by local intravaginal administration of microencapsulated interleukin 12. J Infect Dis. 2013;208:1821–9. doi: 10.1093/infdis/jit354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Liu Y, Liu W, Russell MW. Suppression of host adaptive immune responses by Neisseria gonorrhoeae: role of interleukin 10 and type 1 regulatory T cells. Mucosal Immunol. 2014;7:165–76. doi: 10.1038/mi.2013.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Liu Y, Russell MW. Diversion of the immune response to Neisseria gonorrhoeae from Th17 to Th1/Th2 by treatment with anti-transforming growth factor beta antibody generates immunological memory and protective immunity. MBio. 2011;2:e00095–11. doi: 10.1128/mBio.00095-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Lorenzen DR, Gunther D, Pandit J, et al. Neisseria gonorrhoeae porin modifies the oxidative burst of human professional phagocytes. Infect Immun. 2000;68:6215–22. doi: 10.1128/iai.68.11.6215-6222.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Mandrell RE, Apicella MA. Lipo-oligosaccharides (LOS) of mucosal pathogens: molecular mimicry and host-modification of LOS. Immunobiology. 1993;187:382–402. doi: 10.1016/S0171-2985(11)80352-9. [DOI] [PubMed] [Google Scholar]
  92. Martin D, Cadieux N, Hamel J, Brodeur BR. Highly conserved Neisseria meningitidis surface protein confers protection against experimental infection. J Exp Med. 1997;185:1173–83. doi: 10.1084/jem.185.7.1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Martin I, Sawatzky P, Allen V, et al. Emergence and characterization of Neisseria gonorrhoeae isolates with decreased susceptibilities to ceftriaxone and cefixime in Canada: 2001–2010. Sex Transm Dis. 2012;39:316–23. doi: 10.1097/OLQ.0b013e3182401b69. [DOI] [PubMed] [Google Scholar]
  94. Mascola JR, Stiegler G, VanCott TC, et al. Protection of macaques against vaginal transmission of a pathogenic HIV-1/SIV chimeric virus by passive infusion of neutralizing antibodies. Nat Med. 2000;6:207–10. doi: 10.1038/72318. [DOI] [PubMed] [Google Scholar]
  95. Massari P, Ram S, Macleod H, Wetzler LM. The role of porins in neisserial pathogenesis and immunity. Trends Microbiol. 2003;11:87–93. doi: 10.1016/s0966-842x(02)00037-9. [DOI] [PubMed] [Google Scholar]
  96. McGee ZA, Johnson AP, Taylor-Robinson D. Pathogenic mechanisms of Neisseria gonorrhoeae: observations on damage to human fallopian tubes in organ culture by gonococci of colony type 1 or type 4. J Infect Dis. 1981;143:413–22. doi: 10.1093/infdis/143.3.413. [DOI] [PubMed] [Google Scholar]
  97. McNamara LA, Shumate AM, Johnsen P, et al. First use of a serogroup B meningococcal vaccine in the US in response to a university outbreak. Pediatrics. 2015;135:798–804. doi: 10.1542/peds.2014-4015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. McQuillen DP, Gulati S, Ram S, et al. Complement processing and immunoglobulin binding to Neisseria gonorrhoeae determined in vitro simulates in vivo effects. J Infect Dis. 1999;179:124–35. doi: 10.1086/314545. [DOI] [PubMed] [Google Scholar]
  99. Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004;172:2731–8. doi: 10.4049/jimmunol.172.5.2731. [DOI] [PubMed] [Google Scholar]
  100. Mickelsen PA, Blackman E, Sparling PF. Ability of Neisseria gonorrhoeae, Neisseria meningitidis, and commensal Neisseria species to obtain iron from lactoferrin. Infect Immun. 1982;35:915–20. doi: 10.1128/iai.35.3.915-920.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Mietzner TA, Thomas CE, Hobbs MM, Cohen MS. Vaccines against gonoccoal infection. In: Levine MM, Kaper JB, Rappouli R, et al., editors. New generation vaccines, 3rd ed. Revised and Expanded. New York: Marcel Dekker Inc.; 2004. pp. 755–73. [Google Scholar]
  102. Morales P, Reyes P, Vargas M, et al. Infection of human fallopian tube epithelial cells with Neisseria gonorrhoeae protects cells from tumor necrosis factor alpha-induced apoptosis. Infect Immun. 2006;74:3643–50. doi: 10.1128/IAI.00012-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Mosleh IM, Huber LA, Steinlein P, et al. Neisseria gonorrhoeae porin modulates phagosome maturation. J Biol Chem. 1998;273:35332–8. doi: 10.1074/jbc.273.52.35332. [DOI] [PubMed] [Google Scholar]
  104. Naz RK. Female genital tract immunity: distinct immunological challenges for vaccine development. J Reprod Immunol. 2012;93:1–8. doi: 10.1016/j.jri.2011.09.005. [DOI] [PubMed] [Google Scholar]
  105. Ngampasutadol J, Ram S, Blom AM, et al. Human C4b-binding protein selectively interacts with Neisseria gonorrhoeae and results in species-specific infection. Proc Natl Acad Sci USA. 2005;102:17142–7. doi: 10.1073/pnas.0506471102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Ngampasutadol J, Ram S, Gulati S, et al. Human factor H interacts selectively with Neisseria gonorrhoeae and results in species-specific complement evasion. J Immunol. 2008a;180:3426–35. doi: 10.4049/jimmunol.180.5.3426. [DOI] [PubMed] [Google Scholar]
  107. Ngampasutadol J, Rice PA, Walsh MT, Gulati S. Characterization of a peptide vaccine candidate mimicking an oligosaccharide epitope of Neisseria gonorrhoeae and resultant immune responses and function. Vaccine. 2006;24:157–70. doi: 10.1016/j.vaccine.2005.07.065. [DOI] [PubMed] [Google Scholar]
  108. Ngampasutadol J, Tran C, Gulati S, et al. Species-specificity of Neisseria gonorrhoeae infection: do human complement regulators contribute? Vaccine. 2008b;26:I62–6. doi: 10.1016/j.vaccine.2008.11.051. [DOI] [PubMed] [Google Scholar]
  109. NNDSS. (2015). National notifiable diseases surveillance system. Australian Government Department of Health. [On-line]. Available from: http://www9.health.gov.au/cda/source/rpt_3_sel.cfm [last accessed 1 Oct 2015]
  110. Ober WB. Boswell's clap. JAMA. 1970;212:91–5. [PubMed] [Google Scholar]
  111. Ohnishi M, Golparian D, Shimuta K, et al. Is Neisseria gonorrhoeae initiating a future era of untreatable gonorrhea? Detailed characterization of the first strain with high-level resistance to ceftriaxone. Antimicrob Agents Chemother. 2011;55:3538–45. doi: 10.1128/AAC.00325-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. O'Reilly RJ, Lee L, Welch BG. Secretory IgA antibody responses to Neisseria gonorrhoeae in the genital secretions of infected females. J Infect Dis. 1976;133:113–25. doi: 10.1093/infdis/133.2.113. [DOI] [PubMed] [Google Scholar]
  113. Owusu-Edusei K, Jr, Chesson HW, Gift TL, et al. The estimated direct medical cost of selected sexually transmitted infections in the United States, 2008. Sex Transm Dis. 2013;40:197–201. doi: 10.1097/OLQ.0b013e318285c6d2. [DOI] [PubMed] [Google Scholar]
  114. Packiam M, Shell DM, Liu SV, et al. Differential expression and transcriptional analysis of the alpha-2,3-sialyltransferase gene in pathogenic Neisseria spp. Infect Immun. 2006;74:2637–50. doi: 10.1128/IAI.74.5.2637-2650.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Packiam M, Veit SJ, Anderson DJ, et al. Mouse strain-dependent differences in susceptibility to Neisseria gonorrhoeae infection and induction of innate immune responses. Infect Immun. 2010;78:433–40. doi: 10.1128/IAI.00711-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Papp JR, Schachter J, Gaydos CA, et al. Recommendations for the laboratory-based detection of Chlamydia trachomatis and Neisseria gonorrhoeae-2014. MMWR Recomm Rep. 2014;63:1–19. [PMC free article] [PubMed] [Google Scholar]
  117. Parr MB, Parr EL. Langerhans cells and T lymphocyte subsets in the murine vagina and cervix. Biol Reprod. 1991;44:491–8. doi: 10.1095/biolreprod44.3.491. [DOI] [PubMed] [Google Scholar]
  118. Pettersson A, Kortekaas J, Weynants VE, et al. Vaccine potential of the Neisseria meningitidis lactoferrin-binding proteins LbpA and LbpB. Vaccine. 2006;24:3545–57. doi: 10.1016/j.vaccine.2006.02.003. [DOI] [PubMed] [Google Scholar]
  119. Pettit RK, Judd RC. The interaction of naturally elaborated blebs from serum-susceptible and serum-resistant strains of Neisseria gonorrhoeae with normal human serum. Mol Microbiol. 1992;6:729–34. doi: 10.1111/j.1365-2958.1992.tb01522.x. [DOI] [PubMed] [Google Scholar]
  120. Pound P, Ebrahim S, Sandercock P, et al. Where is the evidence that animal research benefits humans? BMJ. 2004;328:514–17. doi: 10.1136/bmj.328.7438.514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Price GA, Masri HP, Hollander AM, et al. Gonococcal transferrin binding protein chimeras induce bactericidal and growth inhibitory antibodies in mice. Vaccine. 2007;25:7247–60. doi: 10.1016/j.vaccine.2007.07.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Pudney J, Quayle AJ, Anderson DJ. Immunological microenvironments in the human vagina and cervix: mediators of cellular immunity are concentrated in the cervical transformation zone. Biol Reprod. 2005;73:1253–63. doi: 10.1095/biolreprod.105.043133. [DOI] [PubMed] [Google Scholar]
  123. Ram S, Cullinane M, Blom AM, et al. Binding of C4b-binding protein to porin: a molecular mechanism of serum resistance of Neisseria gonorrhoeae . J Exp Med. 2001;193:281–95. doi: 10.1084/jem.193.3.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Ramsey KH, Schneider H, Cross AS, et al. Inflammatory cytokines produced in response to experimental human gonorrhea. J Infect Dis. 1995;172:186–91. doi: 10.1093/infdis/172.1.186. [DOI] [PubMed] [Google Scholar]
  125. Ramsey KH, Schneider H, Kuschner RA, et al. Inflammatory cytokine response to experimental human infection with Neisseria gonorrhoeae . Ann N Y Acad Sci. 1994;730:322–5. doi: 10.1111/j.1749-6632.1994.tb44280.x. [DOI] [PubMed] [Google Scholar]
  126. Rappuoli R. Bridging the knowledge gaps in vaccine design. Nat Biotechnol. 2007;25:1361–6. doi: 10.1038/nbt1207-1361. [DOI] [PubMed] [Google Scholar]
  127. Rechner C, Kuhlewein C, Muller A, et al. Host glycoprotein Gp96 and scavenger receptor SREC interact with PorB of disseminating Neisseria gonorrhoeae in an epithelial invasion pathway. Cell Host Microbe. 2007;2:393–403. doi: 10.1016/j.chom.2007.11.002. [DOI] [PubMed] [Google Scholar]
  128. Rendi MH, Muehlenbachs A, Garcia RL, Boyd KL. Female reproductive system. In: Treuting PM, Dintzis SM, editors. Comparative anatomy and histology: a mouse and human atlas. London, UK: Academic Press; 2012. pp. 253–84. [Google Scholar]
  129. Rice PA, Vayo HE, Tam MR, Blake MS. Immunoglobulin G antibodies directed against protein III block killing of serum-resistant Neisseria gonorrhoeae by immune serum. J Exp Med. 1986;164:1735–48. doi: 10.1084/jem.164.5.1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Rinaudo CD, Telford JL, Rappuoli R, Seib KL. Vaccinology in the genome era. J Clin Invest. 2009;119:2515–25. doi: 10.1172/JCI38330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Ross JD, Moyes A, Young H. Serovar specific immunity to Neisseria gonorrhoeae: does it exist? Genitourin Med. 1995;71:367–9. doi: 10.1136/sti.71.6.367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Rothbard JB, Fernandez R, Wang L, et al. Antibodies to peptides corresponding to a conserved sequence of gonococcal pilins block bacterial adhesion. Proc Natl Acad Sci USA. 1985;82:915–19. doi: 10.1073/pnas.82.3.915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Rotman E, Seifert HS. The genetics of Neisseria species. Annu Rev Genet. 2014;48:405–31. doi: 10.1146/annurev-genet-120213-092007. [DOI] [PubMed] [Google Scholar]
  134. Sadarangani M, Pollard AJ, Gray-Owen SD. Opa proteins and CEACAMs: pathways of immune engagement for pathogenic Neisseria. FEMS Microbiol Rev. 2011;35:498–514. doi: 10.1111/j.1574-6976.2010.00260.x. [DOI] [PubMed] [Google Scholar]
  135. Saigh JH, Sanders CC, Sanders WE., Jr Inhibition of Neisseria gonorrhoeae by aerobic and facultatively anaerobic components of the endocervical flora: evidence for a protective effect against infection. Infect Immun. 1978;19:704–10. doi: 10.1128/iai.19.2.704-710.1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Santin AD, Bellone S, Roman JJ, et al. Trastuzumab treatment in patients with advanced or recurrent endometrial carcinoma overexpressing HER2/neu. Int J Gynaecol Obstet. 2008;102:128–31. doi: 10.1016/j.ijgo.2008.04.008. [DOI] [PubMed] [Google Scholar]
  137. Schmidt KA, Schneider H, Lindstrom JA, et al. Experimental gonococcal urethritis and reinfection with homologous gonococci in male volunteers. Sex Transm Dis. 2001;28:555–64. doi: 10.1097/00007435-200110000-00001. [DOI] [PubMed] [Google Scholar]
  138. Schneider H, Griffiss JM, Boslego JW, et al. Expression of paragloboside-like lipooligosaccharides may be a necessary component of gonococcal pathogenesis in men. J Exp Med. 1991;174:1601–5. doi: 10.1084/jem.174.6.1601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Schoolnik GK, Tai JY, Gotschlich EC. A pilus peptide vaccine for the prevention of gonorrhea. Prog Allergy. 1983;33:314–31. [PubMed] [Google Scholar]
  140. Seib KL, Rappuoli R. Difficulties in developing neisserial vaccines. In: Genco CA, Wetzler LM, editors. Neisseria: molecular mechanisms of pathogenesis. Norwich: Horizon Scientific Press; 2010. pp. 195–226. [Google Scholar]
  141. Seib KL, Scarselli M, Comanducci M, et al. Neisseria meningitidis factor H-binding protein fHbp: a key virulence factor and vaccine antigen. Expert Rev Vaccines. 2015;14:841–59. doi: 10.1586/14760584.2015.1016915. [DOI] [PubMed] [Google Scholar]
  142. Seib KL, Simons MP, Wu HJ, et al. Investigation of oxidative stress defenses of Neisseria gonorrhoeae by using a human polymorphonuclear leukocyte survival assay. Infect Immun. 2005;73:5269–72. doi: 10.1128/IAI.73.8.5269-5272.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Seifert HS, Wright CJ, Jerse AE, et al. Multiple gonococcal pilin antigenic variants are produced during experimental human infections. J Clin Invest. 1994;93:2744–9. doi: 10.1172/JCI117290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Seok J, Warren HS, Cuenca AG, et al. Inflammation, Host Response to Injury LSCRP Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci USA. 2013;110:3507–12. doi: 10.1073/pnas.1222878110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Serino L, Nesta B, Leuzzi R, et al. Identification of a new OmpA-like protein in Neisseria gonorrhoeae involved in the binding to human epithelial cells and in vivo colonization. Mol Microbiol. 2007;64:1391–403. doi: 10.1111/j.1365-2958.2007.05745.x. [DOI] [PubMed] [Google Scholar]
  146. Shell DM, Chiles L, Judd RC, et al. The Neisseria lipooligosaccharide-specific alpha-2,3-sialyltransferase is a surface-exposed outer membrane protein. Infect Immun. 2002;70:3744–51. doi: 10.1128/IAI.70.7.3744-3751.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Shewell LK, Ku SC, Schulz BL, et al. Recombinant truncated AniA of pathogenic Neisseria elicits a non-native immune response and functional blocking antibodies. Biochem Biophys Res Commun. 2013;431:215–20. doi: 10.1016/j.bbrc.2012.12.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Siegel M, Olsen D, Critchlow C, Buchanan TM. Gonococcal pili: safety and immunogenicity in humans and antibody function in vitro. J Infect Dis. 1982;145:300–10. doi: 10.1093/infdis/145.3.300. [DOI] [PubMed] [Google Scholar]
  149. Simons MP, Nauseef WM, Apicella MA. Interactions of Neisseria gonorrhoeae with adherent polymorphonuclear leukocytes. Infect Immun. 2005;73:1971–7. doi: 10.1128/IAI.73.4.1971-1977.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Simons MP, Nauseef WM, Griffith TS, Apicella MA. Neisseria gonorrhoeae delays the onset of apoptosis in polymorphonuclear leukocytes. Cell Microbiol. 2006;8:1780–90. doi: 10.1111/j.1462-5822.2006.00748.x. [DOI] [PubMed] [Google Scholar]
  151. Smith H, Parsons NJ, Cole JA. Sialylation of neisserial lipopolysaccharide: a major influence on pathogenicity. Microb Pathog. 1995;19:365–77. doi: 10.1006/mpat.1995.0071. [DOI] [PubMed] [Google Scholar]
  152. Stephens DS. Gonococcal and meningococc pathogenesis as defined by human cell, cell culture, and organ culture assays. Clin Microbiol Rev. 1989;2:S104–11. doi: 10.1128/cmr.2.suppl.s104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Stohl EA, Criss AK, Seifert HS. The transcriptome response of Neisseria gonorrhoeae to hydrogen peroxide reveals genes with previously uncharacterized roles in oxidative damage protection. Mol Microbiol. 2005;58:520–32. doi: 10.1111/j.1365-2958.2005.04839.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Stohl EA, Dale EM, Criss AK, Seifert HS. Neisseria gonorrhoeae metalloprotease NGO1686 is required for full piliation, and piliation is required for resistance to H2O2- and neutrophil-mediated killing. MBio. 2013;4:e00399–13. doi: 10.1128/mBio.00399-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Stork M, Bos MP, Jongerius I, et al. An outer membrane receptor of Neisseria meningitidis involved in zinc acquisition with vaccine potential. PLoS Pathog. 2010;6:e1000969. doi: 10.1371/journal.ppat.1000969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Su CF, Tsai HJ, Kuo C, et al. Primary non-Hodgkin's lymphoma of the uterus, cervix and parametrium treated by combined immunochemotherapy. J Obstet Gynaecol Res. 2008;34:749–53. doi: 10.1111/j.1447-0756.2008.00919.x. [DOI] [PubMed] [Google Scholar]
  157. Swanson J, Barrera O, Sola J, Boslego J. Expression of outer membrane protein II by gonococci in experimental gonorrhea. J Exp Med. 1988;168:2121–9. doi: 10.1084/jem.168.6.2121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Swanson J, Robbins K, Barrera O, et al. Gonococcal pilin variants in experimental gonorrhea. J Exp Med. 1987;165:1344–57. doi: 10.1084/jem.165.5.1344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Tapsall JW, Ndowa F, Lewis DA, Unemo M. Meeting the public health challenge of multidrug- and extensively drug-resistant Neisseria gonorrhoeae . Expert Rev Anti Infect Ther. 2009;7:821–34. doi: 10.1586/eri.09.63. [DOI] [PubMed] [Google Scholar]
  160. Timmerman MM, Shao JQ, Apicella MA. Ultrastructural analysis of the pathogenesis of Neisseria gonorrhoeae endometrial infection. Cell Microbiol. 2005;7:627–36. doi: 10.1111/j.1462-5822.2005.00491.x. [DOI] [PubMed] [Google Scholar]
  161. Tramont EC, Sadoff JC, Boslego JW, et al. Gonococcal pilus vaccine. Studies of antigenicity and inhibition of attachment. J Clin Invest. 1981;68:881–8. doi: 10.1172/JCI110343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Tramont EC. Inhibition of adherence of Neisseria gonorrhoeae by human genital secretions. J Clin Invest. 1977;59:117–24. doi: 10.1172/JCI108608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Tramont EC. Gonococcal vaccines. Clin Microbiol Rev. 1989;2:S74–7. doi: 10.1128/cmr.2.suppl.s74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Tramont EC, Ciak J, Boslego J, et al. (1980). Antigenic specificity of antibodies in vaginal secretions during infection with Neisseria gonorrhoeae. J Infect Dis 142:23–31 [DOI] [PubMed]
  165. Unemo M, Golparian D, Nicholas R, et al. High-level cefixime- and ceftriaxone-resistant Neisseria gonorrhoeae in France: novel penA mosaic allele in a successful international clone causes treatment failure. Antimicrob Agents Chemother. 2012;56:1273–80. doi: 10.1128/AAC.05760-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Unemo M, Golparian D, Stary A, Eigentler A. First Neisseria gonorrhoeae strain with resistance to cefixime causing gonorrhoea treatment failure in Austria, 2011. Euro Surveill. 2011;16:19998–20000. [PubMed] [Google Scholar]
  167. Unemo M, Nicholas RA. Emergence of multidrug-resistant, extensively drug-resistant and untreatable gonorrhea. Future Microbiol. 2012;7:1401–22. doi: 10.2217/fmb.12.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Unemo M, Shafer WM. Antimicrobial resistance in Neisseria gonorrhoeae in the 21st century: past, evolution, and future. Clin Microbiol Rev. 2014;27:587–13. doi: 10.1128/CMR.00010-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Virji M, Heckels JE. The role of common and type-specific pilus antigenic domains in adhesion and virulence of gonococci for human epithelial cells. J Gen Microbiol. 1984;130:1089–95. doi: 10.1099/00221287-130-5-1089. [DOI] [PubMed] [Google Scholar]
  170. Virji M, Watt SM, Barker S, et al. The N-domain of the human CD66a adhesion molecule is a target for Opa proteins of Neisseria meningitidis and Neisseria gonorrhoeae . Mol Microbiol. 1996;22:929–39. doi: 10.1046/j.1365-2958.1996.01548.x. [DOI] [PubMed] [Google Scholar]
  171. Warner DM, Folster JP, Shafer WM, Jerse AE. Regulation of the MtrC-MtrD-MtrE efflux-pump system modulates the in vivo fitness of Neisseria gonorrhoeae . J Infect Dis. 2007;196:1804–12. doi: 10.1086/522964. [DOI] [PubMed] [Google Scholar]
  172. Warner DM, Shafer WM, Jerse AE. Clinically relevant mutations that cause derepression of the Neisseria gonorrhoeae MtrC-MtrD-MtrE efflux pump system confer different levels of antimicrobial resistance and in vivo fitness. Mol Microbiol. 2008;70:462–78. doi: 10.1111/j.1365-2958.2008.06424.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Welch BG, O'Reilly RJ. An indirect fluorescent-antibody technique for study of uncomplicated gonorrhea. I. Methodology. J Infect Dis. 1973;127:69–76. doi: 10.1093/infdis/127.1.69. [DOI] [PubMed] [Google Scholar]
  174. WHO. (2011). Prevalence and incidence of selected sexually transmitted infections. Chlamydia trachomatis, Neisseria gonorrhoeae, syphilis and Trichomonas vaginalis. Methods and results used by WHO to generate 2005 estimates. on World Health Organisation. [On-line]. Available from http://www.who.int/reproductivehealth/publications/rtis/9789241502450/en/ [last accessed 7 Mar 2014]
  175. WHO. (2012). Global action plan to control the spread and impact of antimicrobial resistance in Neisseria gonorrhoeae (2012), on World Health Organization (WHO), Department of Reproductive Health and Research. [On-line]. Available from: http://www.who.int/reproductivehealth/publications/rtis/9789241503501/en/ [last accessed 7 Mar 2014]
  176. Yeaman GR, Collins JE, Fanger MW, et al. CD8+ T cells in human uterine endometrial lymphoid aggregates: evidence for accumulation of cells by trafficking. Immunology. 2001;102:434–40. doi: 10.1046/j.1365-2567.2001.01199.x. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Critical Reviews in Microbiology are provided here courtesy of Taylor & Francis

RESOURCES