- Open access
- Published: 11 November 2021
Gonorrhoea: a systematic review of prevalence reporting globally
- Jane Whelan ORCID: orcid.org/0000-0003-3784-2749 1 ,
- Victoria Abbing-Karahagopian ORCID: orcid.org/0000-0003-1987-363X 1 ,
- Laura Serino ORCID: orcid.org/0000-0002-8618-9726 2 &
- Magnus Unemo ORCID: orcid.org/0000-0003-1710-2081 3
BMC Infectious Diseases volume 21 , Article number: 1152 ( 2021 ) Cite this article
15k Accesses
33 Citations
3 Altmetric
Metrics details
The World Health Organization (WHO) recommends periodic gonorrhoea prevalence assessments in the general population or proxies thereof (including pregnant women, women attending family planning clinics, military recruits, and men undergoing employment physicals for example) and in population groups at increased risk, including men-who-have-sex-with-men (MSM) and sex workers.
We evaluated reported prevalence data, including estimates from proxy general population samples to reflect the WHO recommendations. We describe the outcomes from the general population country-by-country and extend previous reviews to include MSM, sex workers, and extragenital infections.
Result and conclusion
In our systematic search, 2015 titles were reviewed (January 2010–April 2019) and 174 full-text publications were included. National, population-based prevalence data were identified in only four countries (the United States of America, the United Kingdom, Peru, New Caledonia) and local population-based estimates were reported in areas within five countries (China, South Africa, Brazil, Benin, and Malawi). The remaining studies identified only reported test positivity from non-probability, proxy general population samples. Due to the diversity of the reviewed studies, detailed comparison across studies was not possible. In MSM, data were identified from 64 studies in 25 countries. Rectal infection rates were generally higher than urogenital or pharyngeal infection rates, where extragenital testing was conducted. Data on sex workers were identified from 41 studies in 23 countries; rates in female sex workers were high. Current prevalence monitoring was shown to be highly suboptimal worldwide. Serial prevalence monitoring of critical epidemiological variables, and guidelines to optimize prevalence study conduct and reporting beyond antenatal settings are recommended.
Peer Review reports
Gonorrhoea is a sexually transmitted infection (STI) caused by Neisseria gonorrhoeae (the gonococcus). In 2016, an estimated 87 million incident cases occurred among persons aged 15–49 years worldwide with an incidence rate of 20 cases/1000 women and 26/1000 men [ 1 ].
Gonorrhoea affects the urogenital tract, oropharynx, rectum, or conjunctiva, and repeat infections are common. Urogenital infections are often asymptomatic, particularly in women, but irrespective of symptoms, gonorrhoea is associated with substantial morbidity. Serious complications and sequelae include pelvic inflammatory disease, chronic pelvic pain, ectopic pregnancy, and infertility in women [ 2 ]. Infection during pregnancy is also associated with low birth weight and neonatal conjunctivitis, which can progress to blindness [ 2 , 3 ]. In men, gonorrhoea can cause epididymitis [ 2 ]. Rectal and pharyngeal gonorrhoea cases, mostly asymptomatic, are prevalent in men-who-have-sex-with-men (MSM), but can be common also in women and, particularly pharyngeal infection, in men who have sex only with women [ 4 ]. The presence of gonorrhoea is also a co-factor in human immunodeficiency virus (HIV) transmission [ 5 ].
Gonorrhoea is substantially underdiagnosed and underreported worldwide [ 3 ]. Even in high-income economies with well-established STI surveillance systems, it is estimated that more than half of infections are unidentified or unreported [ 6 , 7 ]. This underdiagnosis/underreporting is higher in less-resourced settings and settings using syndromic management with limited access to state-of-the-art diagnostics such as nucleic acid amplification tests (NAATs). Though partially explained by the asymptomatic nature of the infection, underreporting is also due to delays in seeking healthcare and inaccessible or inadequate STI testing/treatment in underserved populations or those particularly vulnerable to infection: adolescents and young people, some ethnic and racial groups, communities of lower socioeconomic status, MSM, sex workers, and others [ 8 ].
The World Health Organization (WHO)’s global target is a 90% reduction in gonorrhoea cases by 2030 [ 9 ]. To monitor progress towards this goal, STI trend monitoring at the national level is recommended. This should include routine prevalence assessments (every two to three years) of bacterial STIs among general populations of men and women (e.g. including pregnant women, women attending family planning clinics, military recruits and men undergoing employment physicals) [ 3 ]. Monitoring in high-risk priority populations including MSM and sex workers is also recommended [ 3 , 9 ].
The WHO reports prevalence estimates of curable non-viral STIs at a global and regional level using epidemic models, while recognizing the small number of prevalence data points that are available to generate reliable estimate [ 1 , 3 ]. Notably, for key population groups such as MSM and sex workers, who likely contribute substantially to the worldwide infection burden, gonorrhoea prevalence in global estimates is indirectly accounted for [ 1 ] and estimates do not reflect rectal and pharyngeal infection.
N. gonorrhoeae is progressively developing antimicrobial resistance (AMR) to all therapeutic antibiotics, and the WHO has issued warnings that untreatable gonorrhoea may be on the horizon [ 10 ]. National prevalence estimates are an essential indicator of the state of gonorrhoea and STI control at state level and globally [ 3 ]. In this review, we aimed to evaluate global prevalence reporting in the general population, and proxies thereof, on a country-by-country basis, extending previous reviews to report on key population groups of MSM and female and male sex workers (FSW and MSW), including extragenital as well as urogenital infection.
Search strategy and selection criteria
We conducted a systematic search of PubMed following PRISMA guidelines (Additional file 1 ) for papers published from 1 January 2010 to 11 April 2019. We derived a sensitive search strategy requiring at least one medical subject headings (MeSH) term related to a sexually transmitted disease (STD) or gonorrhoea and at least one reference to the keyword ‘gonorrhoea’ in the title or abstract. We did not specify the population (e.g. MSM, FSW or MSW), as we noted substantial overlap in reporting of risk groups and inclusion of terms such as ‘prevalence’, ‘epidemiology’ or ‘rate’ rendered the search too specific, omitting relevant papers (Additional file 2 ). Two authors (JW and VAK) independently screened all titles and abstracts against pre-specified inclusion and exclusion criteria (Additional file 3 ) and agreed on the selection of articles to be obtained as full text. English-language abstracts were reviewed but the full text was translated as necessary, from Portuguese, Spanish, and Chinese, where relevant. The systematic search was supplemented with an online English-language country-by-country search of websites, data repositories and surveillance reports of public health and/or governmental agencies using the country name, and ‘gonorr*’ or ‘sexually transmitted’ and ‘disease’ or ‘infection’ to identify data sources and provide context to prevalence estimates. We reviewed regional and international health agency data (WHO, European Centre for Disease Prevention and Control [ECDC]) and contacted relevant experts in the field. AMR monitoring, an essential component of gonorrhoea surveillance [ 10 ] and worthy of a separate review, was beyond the scope of this search.
Data analysis
The primary outcome (prevalence of gonorrhoea) was defined as the proportion of persons with laboratory-confirmed (culture and/or NAAT positive) gonorrhoea in the population within a specified time. It became apparent early in the literature search that population-based prevalence estimates were very limited and so to address the WHO recommendation to derive estimates from studies which are not necessarily population-based but nevertheless relevant, we defined a post-hoc secondary objective to report test positivity, categorizing these as proxy general population samples. Data were tabulated by population group (classified as ‘general population’, MSM and sex workers) and summarized per WHO region and country. ‘General population’ samples were identified according to WHO recommendations, to include studies conducted ‘among pregnant women, women attending family planning clinics, male military recruits and men undergoing employment physicals’ [ 3 ]. These samples served as proxies for the general population where population-based sampling was not, or could not, be conducted. The point estimates reported were adjusted for diagnostic test performance by applying a standardization factor for urogenital infection as utilized by WHO (Additional file 4 ) [ 3 , 11 ]. For rectal and pharyngeal infections, a separate literature review was undertaken to derive sensitivity and specificity values (for culture and/or NAAT) and adjustments were applied in the same manner as for the urogenital samples (Additional file 4 ). Due to obvious heterogeneity in study populations and study designs, widespread inclusion of non-representative samples and frequent lack of reporting of key parameters to judge the study quality, a quality score was not assigned. Similarly, a meta-analysis could not be conducted as we were limited in our ability to appropriately compare studies directly. We did not calculate a median summary estimate per country because only a small number of countries had three or more available estimates. Instead, guided by the principles of Campbell et al [ 12 ], we conducted a narrative synthesis, presenting the prevalence and test positivity estimates reported in the context of the source population and the type of sampling conducted, rather than directly comparing estimates. General population estimates were considered ‘population-based’ and representative if participants were sampled from a general population sampling frame and some form of random selection was performed. Studies employing other forms of sampling from proxy general population samples are labelled as such. As MSM and sex workers are defined in terms of their sexual behaviour, population-based denominator samples are generally not available. For these groups, screening and/or enhanced testing is frequently recommended irrespective of symptom status (and thus may be more reflective of prevalence). Therefore, studies conducted at STI clinics and at other venues frequented by MSM and sex workers were eligible for inclusion, excepting studies including persons presenting with symptoms, which were excluded to minimize bias. The median sample size and interquartile range were estimated using Excel’s ‘quartile.exc’ function.
Prevalence reporting in the general population
We identified 2015 citations relating to gonorrhoea ‘prevalence’ (Fig. 1 ), subsequently categorized into (a) the general population or proxy general population groups (men, women, and pregnant women separately), (b) MSM, and (c) FSW and MSW. Following title and abstract screening, we reviewed 424 full-text publications, of which 174 addressed the primary or secondary objective and were eligible for inclusion, reporting data from the following WHO regions: Africa (n=41), the Western Pacific (n=41), high-income North America that is part of the Region of the Americas (n=25), the Americas excluding high-income North America (n=25), Europe (n=19), South-East Asia (n=18), and the Eastern Mediterranean (n=5). The number of countries where prevalence and/or test positivity estimates were identified from the general population was limited, with data points identified from only 18.0% of countries worldwide (35/194) for women and 9.8% (19/194) for men (Fig. 2 ). Prevalence of gonorrhoea in the general population by WHO region and country is summarized in Table 1 and test positivity estimates from proxy general population samples in Table 2 .
PRISMA diagram describing selection of citations reporting gonorrhoea prevalence. Note: Some articles reported outcomes on several of the populations of interest or provided data for >1 country and therefore the total number of included data points does not amount to 174. n=number of articles
Availability of gonorrhoea prevalence reporting globally. Maps represent the availability of prevalence data in general population samples worldwide, including pregnant women, women attending family planning clinics, male military recruits, and work-based health screening programmes and other similar groups. General population estimates were considered ‘national population-based’ or ‘local population-based’ if participants were sampled from a general population sampling frame and some form of random selection was performed. Studies where probability sampling was not conducted, and which may not be generalizable beyond the study, are labelled as ‘non-population based’
For several countries, we did not identify prevalence or test positivity data. The grey literature search led to one additional estimate [ 63 ], but also allowed us to set the prevalence estimates identified in the context of the extent of surveillance otherwise ongoing in the country. To this end, expert consultation led to identification of surveillance data from three international reporting networks (WHO Global, WHO European Regional Office, and ECDC), and national surveillance data or reports from an additional seven countries, the United States of America (USA), Canada, Australia, Singapore, New Zealand, Japan, and the Republic of Korea.
Prevalence data and/or test positivity in general population samples were identified in 13.2% (seven out of 53) of countries in the WHO European region (nine estimates in women, including pregnant women [ 18 , 55 , 56 , 57 , 58 , 59 , 60 , 61 , 62 ], and five in men [ 18 , 56 , 57 , 58 , 59 ]) (Tables 1 and 2 ). We identified only one representative, population-based prevalence study in the United Kingdom (UK) that was of national scope [ 18 ]. These data were derived from the National Survey of Sexual Attitudes and Lifestyles (NATSAL) in 2010–2012. A probability sample of 15 162 men and women aged 16–74 years was drawn from the general population. Gonorrhoea testing was conducted for 2665 women and 1885 men and an overall prevalence of <0.1% was recorded (Table 1 ), higher in women and men aged 20–24 (0.2% and 0.1%, respectively). Data from all other countries represented test positivity data that were drawn from proxy groups of the general population, mainly non-probability samples, drawn from antenatal/obstetric clinics, primary care, community/youth clinics, with one study in a high school setting [ 56 ]. The median study sample size was 1004 in all women (interquartile range [IQR]: 220–5337) and 1236 in men (IQR: 802–6620). In all general population studies, NAAT testing conducted on urine (men, women) or genital fluid (women) was most common; confirmation by both NAAT and culture was used in pregnant women in France and Portugal [ 61 , 62 ]. Data on both sexes were available in only five studies [ 18 , 56 , 57 , 58 , 59 ]. One study reported samples from the urogenital and rectal site in aggregate [ 57 ]. All other studies included urogenital infection only.
For countries where no prevalence or test positivity estimate from the general population was identified, some degree of surveillance data was discoverable through the grey literature search. Most European Union (EU)/European Economic Area (EEA) Member States have comprehensive surveillance systems and report a national notification rate annually, except for Germany, Liechtenstein, Austria (not since 2014), and Greece (not since 2017) [ 85 , 86 ]. Belgium, France, and the Netherlands have sentinel surveillance systems. In countries outside the EU/EAA region (mostly the eastern European region), data were less discoverable. In 2017, countries including Armenia, Azerbaijan, Belarus, Georgia, Kazakhstan, Kyrgyzstan, Russian Federation, Turkmenistan, and Uzbekistan reported gonorrhoea cases to the WHO European Regional Office (M. Dara and G. Kuchukhidze, personal communication, 24 February 2019). Indicators included the absolute number of cases identified, the male to female ratio, and only for Armenia, the proportion of reported MSM among the cases. Prevalence data or comprehensive syndromic and aetiologic case reporting were not otherwise identified in the wider European region.
High-income North America
In the USA, laboratory-confirmed gonorrhoea is mandatorily notifiable and data collection is comprehensive, from diverse clinical settings including STD clinics, laboratories, family planning and school-based clinics, hospitals, emergency rooms, drug treatment centres, correctional facilities, and the military [ 87 ]. The most recent estimate of nationwide population prevalence identified was from the National Health and Nutrition Examination Survey (NHANES), a series of cross-sectional, bi-annual household surveys representative in terms of sex, age and race/ethnicity of the USA civilian, non-institutionalized population [ 19 ]. Between 1999 and 2008, screening for cervical or urethral gonorrhoea was a study component, and 15 885 persons, aged 14–39 years participated. An extrapolated national prevalence of 0.3% (95% confidence interval [CI]: 0.1%–0.5%) among 14–39-year-olds was estimated, higher in women than in men (Table 1 ). N. gonorrhoeae testing within NHANES stopped at the end of 2008 and, in 2009, gonorrhoea prevalence and notification rates were at an all-time low in the USA [ 19 ].
In terms of non-probability samples, an estimate of prevalence from a sentinel surveillance population of young people at elevated risk for gonorrhoea is provided annually by the Centers for Disease Control and Prevention (CDC), using data from the ‘National Job Training Program’ (NJTP), a nationwide vocational programme for socioeconomically disadvantaged youth aged 16 to 24 years who are considered at risk of STIs [ 63 ]. Participants are offered gonorrhoea and chlamydia screening at programme entry. In 2018, the median state-specific estimated gonorrhoea prevalence for programme entrants aged 16–24 years was 2.2% in women (range 0.4% to 7.6%), and 0.7% in men (range 0.0% to 4.8%) (Table 2 ) [ 63 ].
In the USA, we identified a further seven test positivity estimates from proxy general population samples in women (including one from a chart review of women screened [ 64 ], two studies in high schools [ 65 , 66 ], and four in pregnant women who are routinely tested [ 67 , 68 , 69 , 70 ]) and three data points in men (the same two studies in high schools [ 65 , 66 ] and one study in college students [ 81 ]) that met the inclusion criteria for the secondary objective (Table 2 ). There was a wide range in study sample size and in estimates reported, reflecting diversity in study participants and settings, and study population characteristics. Test positivity estimates from non-probability samples from the two studies in high schools were identified: one reported the proportion positive over almost 8 years (9.0% [3270/36 263] in girls and 4.1% [1588/39 010] in boys) and another yielded a combined estimate of 2.4% in girls and boys (Table 2 ) [ 65 , 66 ]. No comparison could be made across studies. Where reported, studies used NAAT testing.
In Canada, no prevalence study or proxy general population study was identified. Gonorrhoea is mandatorily notifiable, and laboratory-confirmed cases are reported to the Public Health Agency of Canada through the Canadian Notifiable Disease Surveillance System. Summary data are published annually by age and sex, and are available online [ 88 ], and a detailed surveillance report is produced every five years.
Americas (excluding high-income North America)
Prevalence and/or test positivity estimates from the general population were identified in 18.2% (six out of 33) of countries in this WHO region excluding the USA and Canada (12 estimates in women, including pregnant women [ 16 , 17 , 44 , 45 , 46 , 47 , 48 , 49 , 50 , 51 , 52 , 53 ], and four in men [ 17 , 48 , 49 , 80 ]) (Tables 1 and 2 ). One study in Peru could be considered population-based and of national scope. In this study, the substantial sample included 13 925 randomly selected 18–29-year-old men and women who were resident in 24 cities with populations >50 000 people [ 17 ]. Additionally, a local population-based study in Brazil, also urban, was conducted using two-stage sampling of households and young women in middle size cities in Central Brazil [ 16 ]. The remaining studies were non-probability samples, mainly from community settings including educational facilities, primary healthcare, adolescent health clinics and ANCs. The median study sample size was 399 in women (IQR: 309-1719) and 371 in men (IQR: 180-5749). All studies involved NAAT screening of urine (n=4) [ 16 , 44 , 45 , 49 ] and urogenital swab samples (n=7) [ 17 , 46 , 47 , 48 , 50 , 51 , 53 ] for women (clinical specimen not specified, n=1 [ 52 ]), and urine (n=3) [ 17 , 48 , 49 ] for men (clinical specimen not specified, n=1 [ 80 ]).
From the grey literature search, we identified only aetiological or syndromic case reporting in adult men through WHO Global AIDS Monitoring (GAM; known as Global AIDS Response Progress Reporting prior to 2015) for other countries in the region [ 3 ]. No further prevalence or test positivity data were identified in the region.
In the WHO African region, prevalence data and/or test positivity estimates from the general population were identified in 25.5% (12 out of 47) of countries (25 estimates in women, including pregnant women [ 13 , 14 , 15 , 23 , 24 , 25 , 26 , 27 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 ], and six in men [ 13 , 14 , 15 , 29 , 31 , 34 ]) (Tables 1 and 2 ). Three of the studies were local population-based, derived from household samples, and none were of national scope. The first was from the urban centre of Cotonou in Benin, where 2507 subjects aged 15–49 years, from 1070 households sampled from 38 census areas, participated [ 13 ]. In Malawi, another estimate was derived from a largely rural population from the eastern lakeside regions of the Mangochi district [ 14 ]. Most recently, 1342 young people aged 15–24 years were selected from a ‘health and demographic surveillance site’ sampling frame in rural South Africa [ 15 ]. The remaining studies (Table 2 ) were derived from non-probability samples with diverse recruitment sites, including antenatal clinic (ANC) settings, schools and universities, primary healthcare sites, and community-based recruitment. The median study sample size was 322 in women (IQR: 200–553) and 422 in men (IQR: 351–755). Laboratory confirmation was mainly by NAAT on urogenital swab samples and, to a lesser extent, on urine for women; in four studies [ 23 , 24 , 30 , 40 ], Gram stain and/or culture only were used. For men, urine samples were tested by NAAT in all cases where reported.
In the African region, 43% of countries reported to WHO in 2013 having STI surveillance systems in place and 40% had national strategies or plans for preventing and controlling STIs [ 89 ], but beyond limited reporting of aetiological surveillance among men and syndromic surveillance in men and women, we did not identify any further prevalence reporting in the region.
Western Pacific
Prevalence data and/or test positivity data from the general population were identified in 22.2% (six out of 27) of countries and territories in the WHO Western Pacific region (11 estimates in women, including pregnant women [ 20 , 21 , 22 , 73 , 74 , 75 , 76 , 77 , 78 , 79 ], and four in men [ 20 , 22 , 73 , 84 ]) (Tables 1 and 2 ). There were three population-based studies. One was of national scope in New Caledonia [ 22 ]. It included men and women selected during a national three-stage random sampling of general practice surgeries and public dispensaries, and the sample was then weighted to reflect the general population aged 18–49 years. The other two population-based studies were local in scope and were both in China. In one study from the Shandong province [ 20 ], men and women were sampled in a complex multi-stage sampling process based on urban and rural communities within geographic regions. The second study, from Shenzhen City [ 21 ], included women only and was designed to be representative of the entire population in the Nanshan District of the city. Beyond these prevalence data, for both men and women, test positivity estimates from non-probability, proxy general population samples were derived from a range of study settings including community settings, primary care, and hospital-based maternity clinics/ANCs. One study was in an occupational group (miners) in men in China [ 84 ]. The median study sample size in the region was 765 in women (IQR: 362–3581) and 1290 in men (IQR: 376–4490). In some countries in the region where no data were identified, gonorrhoea is a notifiable infection; routine national surveillance is conducted and opportunistic/risk-based screening and/or testing is recommended for some population groups (Australia, New Zealand, and Singapore) [ 90 , 91 , 92 ]. Sentinel surveillance is conducted in the Republic of Korea and in Japan, mostly in urology departments. In both countries, reported cases per sentinel are low and have decreased in recent years [ 93 , 94 ]. GAM data for men are also notified to WHO from many countries [ 3 ], but no further prevalence data were identified in the region.
South-East Asia
Among 11 countries in the WHO South-East Asian region, we did not identify any population-based prevalence estimates. Test positivity data in general population samples were identified from 18.2% (two out of eleven) of countries (two estimates in women, including pregnant women [ 71 , 72 ], and two in men [ 82 , 83 ]) (Table 2 ). Non-probability samples from the general population were community-based in women in India, hospital-based in young pregnant women aged <18 years in Thailand, and in occupational groups in men (migrant workers in India and military conscripts in Thailand). The median study sample size was 466 in women (range: 121–811) and 1482 in men (range: 840–2123).
We did not identify further information on gonorrhoea surveillance in the region, with the exception of GAM data from some countries [ 3 ].
Eastern Mediterranean
We identified non-probability samples in 4.8% (one out of 21) of countries in the WHO Eastern Mediterranean region: a single study in a hospital involving pregnant women [ 54 ] (Iran, n=239; standardized prevalence: 0.5%) (Table 2 ). According to the WHO, ten countries surveyed in 2013 reported having an STI surveillance system, four reported conducting aetiological studies, 11 had updated national STI guidelines or recommendations in place and nine had a national strategy or action plan for STI prevention and control [ 89 ], but no further estimates were identified in the region.
Prevalence and test positivity reporting in vulnerable population groups
Men-who-have-sex-with-men
Prevalence and/or non-probability test positivity data on gonorrhoea in the MSM population were identified in 64 studies from 25 countries (seven countries in Africa, five in Europe, two in North America, four in the Americas [excluding high-income North America], four in the Western Pacific, and three in South-East Asia) (Fig. 3 ; Additional file 5 ) [ 95 , 96 , 97 , 98 , 99 , 100 , 101 , 102 , 103 , 104 , 105 , 106 , 107 , 108 , 109 , 110 , 111 , 112 , 113 , 114 , 115 , 116 , 117 , 118 , 119 , 120 , 121 , 122 , 123 , 124 , 125 , 126 , 127 , 128 , 129 , 130 , 131 , 132 , 133 , 134 , 135 , 136 , 137 , 138 , 139 , 140 , 141 , 142 , 143 , 144 , 145 , 146 , 147 ]. For 56.0% (14 out of 25) of countries, data originated from a single study in an urban setting. In five studies, men testing HIV-positive were excluded at the outset [ 96 , 119 , 124 , 148 , 149 ]. HIV status was reported in five studies with variable HIV-positivity [ 98 , 100 , 103 , 115 , 127 ]. Three studies included asymptomatic cases only [ 120 , 131 , 150 ]. Urogenital screening and/or opportunistic testing (predominantly on urine samples) was most often performed. An equal proportion of studies involved recruitment from community settings or STI clinics, but there was diversity in terms of the populations included, including HIV status, which was often not reported. Both rectal and urogenital sampling were reported in 26 studies; rates of rectal infection were higher than urogenital rates in 69.2% (n=18) of these studies (Fig. 3 ). NAAT testing was reported in 22 of these studies, culture-only testing in two, and culture or NAAT testing in two. Reported rates of pharyngeal testing from 27 studies were mostly (51.9%) between 5.0% and 10.0%, 22.2% were between 1.0% and 5.0%, and 14.8% were >10.0%. Though variable, on average, the standardized estimate of pharyngeal infection was similar to urogenital positivity where reported in the same study.
Reported prevalence and/or test positivity of urethral (a) and/or rectal (b) gonorrhoea-positive cases in men-who-have-sex-with-men. 1 Prevalence rates for urethral gonorrhoea could not be standardized. 2 Prevalence rates for rectal gonorrhoea could not be standardized
Sex workers
Data on gonorrhoea prevalence and/or test positivity in MSW, FSW or both were available from 23 countries (Table 3 ), with 38 studies reporting on FSW and six on MSW. Of 41 unique studies, 14 were conducted in a clinic setting (including STI clinics, genito-urinary clinics and outreach clinics) and 13 at commercial sites (including hotels, brothels, street and residence). The remainder (n=14) were described as community-based or conducted at other mixed locations. Only urogenital testing was performed except for one study in China that also performed pharyngeal testing [ 184 ]. Overall, the median study sample size was 655 in women (IQR: 323–2165) and 240 in men (IQR: 113–584). The positivity estimates ranged from 0.0% (MSW in the Republic of Korea) to 29.2% (FSW in Indonesia).
Gonorrhoea prevalence monitoring is one of four key components of national STI surveillance programmes that is recommended by WHO to reduce the burden of gonorrhoea infections by 90% between 2018 and 2030 (in addition to case reporting, assessment of the aetiology of STI syndromes, and monitoring of antimicrobial resistance) [ 9 ]. WHO recommends prevalence assessments in the general population every two to three years, and in key populations such as MSM and sex workers [ 3 , 9 ]. From our review, it is clear that substantive prevalence data among representative samples of the general population were seriously lacking on a worldwide basis. We identified national population-based data from only four countries (USA [ 19 ], UK [ 18 ], Peru [ 17 ], and New Caledonia [ 22 ]), all pre-dating 2013. Recent local population-based data were identified from China [ 20 , 21 ] (2016 and 2017) and South Africa [ 15 ] (2018), but otherwise samples used for local population-based estimates were collected more than 10 years ago (Brazil, Benin, and Malawi [ 13 , 14 , 16 ]). The majority of the remaining test positivity estimates were derived from non-probability samples from groups that might be considered proxies of the general population, as proposed by WHO [ 3 ].
Based on our findings, most studies were conducted in single centres or discrete geographic regions or populations. We excluded STI clinic settings to avoid overestimating the prevalence in general population samples. As estimates (mainly from proxy general population groups) tended to be high, albeit with wide variation in the magnitude and precision of the estimate, it is highly likely that the risk profile of proxy populations was also higher than that of the general population. Even within groups, representativeness may not always have been similar (e.g. military conscripts residing in barracks versus those living at home). The median study sample size in the Americas (excluding high-income North America), Africa and the Eastern Mediterranean was <500 in both men and women, which further limits generalizability beyond the study population in question. Where national data were available to comment (e.g. USA), estimates from non-probability samples in defined younger populations [ 63 , 65 , 66 ] were higher than nationwide population-based estimates [ 19 ] or estimates from older populations [ 64 ], further highlighting the need for continued nationally representative population sampling. Diagnostic testing used varied widely and the sensitivity and specificity of these are an essential factor, contributing to differences in reported estimates. We standardized estimates for differences in laboratory methods (NAAT versus culture) and clinical specimens (urine or urogenital samples) where reported [ 3 , 11 ]. For consistency, we also adjusted for NAAT versus culture on rectal and pharyngeal samples, based on reported sensitivities and specificities in the literature and using a similar standardization procedure, to allow for within-study comparison. The specimen and test were not always reported, but NAAT-based testing was most common.
For most countries, no prevalence estimate or test positivity estimate from general population groups was identified. It was clear from our online (English) grey literature search, that surveillance is ongoing more widely, as we retrieved surveillance reports from online national and international data repositories, syndromic surveillance reports in some countries, and intermittent summaries of laboratory surveillance in others. Generally, the quality and quantity of data identified were highly variable and often neither timely nor contemporaneous. In the absence of prevalence data, low case rates reported in some settings likely reflect limited testing and restricted availability of appropriate laboratory diagnostics rather than actual infection rates. In many African countries, for example, prevalence reports (where available) and syndromic surveillance suggest that the very limited aetiological reporting substantially underestimates the true infection burden.
Heterogenous data in MSM were available for only 12.9% (25 out of 194) of countries, mainly single-centre studies in urban, community-based or STI clinic settings. Most studies performed testing at the urogenital site. Where both urogenital and extragenital testing were conducted, rates at rectal sites were typically higher. However, for modern NAATs no evidence-based consensus exists regarding sensitivity and specificity correction factors when using other diagnostic methods or different NAATs for urogenital and especially extragenital infections. International evidence-based consensus regarding these corrections is imperative to develop. Rates among FSW were often many multiples higher than general population estimates in women, in countries where data in both populations were available. Due to the dearth of data on sex workers in some regions, we erred on the side of inclusivity, including small studies of <100 from Iran for example, where no data were otherwise available.
Our review had limitations. There were undoubtedly data from studies not discoverable on PubMed. For example, a systematic review from China, which documented STI risk among MSM [ 189 ], included studies that we could not access through the library systems available to us. Systematic reviews on a regional basis with good local knowledge, including in setting and language, would be a valuable addition. For many countries, only one or two data points were identified. Limited data and marked heterogeneity between studies prohibited us from conducting a meta-analysis or reporting median estimates. Reporting of proportion testing positive was very diverse in terms of variables reported, degree of stratification by demographic and other factors (e.g. HIV status), details regarding diagnostic tests, and anatomic site, often with statistics omitted where data had clearly been collected. With improved reporting from diverse populations, novel methods for synthesizing diverse data may therefore be required.
Conclusions
Gonorrhoea prevalence is a core indicator to properly inform gonorrhoea management and control programmes, international and national guidelines, and policy documents. Gonorrhoea prevalence monitoring and reporting is suboptimal or absent in most countries. Many countries and regions have seen substantial increases in notification rates of gonorrhoea in recent years [ 63 , 85 ]. In the absence of serial prevalence data, however, it is difficult to disentangle how much of this reflects a true increase in the burden of gonorrhoea or some degree of improved awareness among groups at increased risk (in particular MSM), more consistent screening and/or testing, increased availability and use of NAATs, and improved (electronic) reporting. Irrespectively, among key populations such as MSM and sex workers, there is a substantial burden of infection where data are available. To inform STI control programmes at the national and regional level, and to inform innovative epidemiologic modelling initiatives such as SPECTRUM [ 11 ] and the Global Burden of Disease [ 190 ] that attempt to quantify and model the global burden, significantly more data of higher quality are required. There is an urgent need for more resources for researchers to design, conduct and report prevalence studies in a more consistent, standardized, and quality-assured way. Within countries, serial prevalence monitoring at intervals, including assessment and reporting of a minimum set of epidemiological variables, should be considered. Our review showed the need for more testing at extragenital sites, particularly, but not exclusively, among the MSM population. WHO currently provides guidance on the assessment of gonorrhoea and chlamydia prevalence among pregnant women at ANCs [ 191 ]. This guidance could be extended beyond the ANC setting. Consistent adherence to study reporting guidelines (e.g. adapted STROBE checklists [ 192 ] or equivalent), for all researchers is also advised.
Availability of data and materials
The dataset supporting the conclusions of this article is included within the article and its additional file (Additional file 6 ).
Abbreviations
Antimicrobial resistance
Antenatal clinic
Centers for Disease Control and Prevention
Confidence interval
European Centre for Disease Prevention and Control
European Economic Area
European Union
Female sex workers
Global AIDS Monitoring
Human immunodeficiency virus
Interquartile range
Medical subject headings
Male sex workers
Nucleic acid amplification tests
National Survey of Sexual Attitudes and Lifestyles
National Health and Nutrition Examination Survey
National Job Training Program
Sexually transmitted disease
Sexually transmitted infection
United Kingdom
United States of America
World Health Organization
Rowley J, Vander Hoorn S, Korenromp E, Low N, Unemo M, Abu-Raddad LJ, et al. Chlamydia, gonorrhoea, trichomoniasis and syphilis: global prevalence and incidence estimates, 2016. Bull World Health Organ. 2019;97:548–62P.
Article PubMed PubMed Central Google Scholar
Unemo M, Seifert HS, Hook EW 3rd, Hawkes S, Ndowa F, Dillon JR. Gonorrhoea. Nat Rev Dis Primers. 2019;5:79.
Article PubMed Google Scholar
World Health Organization. Report on global sexually transmitted infection surveillance 2018. https://www.who.int/reproductivehealth/publications/stis-surveillance-2018/en/ . Accessed 9 Oct 2020.
Chan PA, Robinette A, Montgomery M, Almonte A, Cu-Uvin S, Lonks JR, et al. Extragenital infections caused by Chlamydia trachomatis and Neisseria gonorrhoeae: a review of the literature. Infect Dis Obstet Gynecol. 2016;2016:5758387.
Beck EJ, Mandalia S, Leonard K, Griffith RJ, Harris JR, Miller DL. Case-control study of sexually transmitted diseases as cofactors for HIV-1 transmission. Int J STD AIDS. 1996;7:34–8.
Article CAS PubMed Google Scholar
van Lier A, McDonald SA, Bouwknegt M, group EPI, Kretzschmar ME, Havelaar AH, et al. Disease burden of 32 infectious diseases in the Netherlands, 2007-2011. PLoS One. 2016;11:e0153106.
Satterwhite CL, Torrone E, Meites E, Dunne EF, Mahajan R, Ocfemia MC, et al. Sexually transmitted infections among US women and men: prevalence and incidence estimates, 2008. Sex Transm Dis. 2013;40:187–93.
Leichliter JS, Dittus PJ, Copen CE, Aral SO. Trends in factors indicating increased risk for STI among key subpopulations in the United States, 2002-2015. Sex Transm Infect. 2020;96:121–3.
World Health Organization. Global Health Sector Strategy on Sexually Transmitted Infections 2016-2021. https://apps.who.int/iris/bitstream/handle/10665/246296/WHO-RHR-16.09-eng.pdf?sequence=1 . Accessed 7 Oct 2020.
Unemo M, Lahra MM, Cole M, Galarza P, Ndowa F, Martin I, et al. World Health Organization Global Gonococcal Antimicrobial Surveillance Program (WHO GASP): review of new data and evidence to inform international collaborative actions and research efforts. Sex Health. 2019;16:412–25.
Spectrum. Glastonbury: Avenir Health 2019. https://www.avenirhealth.org/software-spectrum.php . Accessed 30 Nov 2020.
Campbell M, McKenzie JE, Sowden A, Katikireddi SV, Brennan SE, Ellis S, et al. Synthesis without meta-analysis (SWiM) in systematic reviews: reporting guideline. BMJ. 2020;368:l6890.
Behanzin L, Diabate S, Minani I, Lowndes CM, Boily MC, Labbe AC, et al. Decline in HIV prevalence among young men in the general population of Cotonou, Benin, 1998-2008. PLoS One. 2012;7:e43818.
Article CAS PubMed PubMed Central Google Scholar
Paz-Soldan VA, Hoffman I, de Graft JJ, Bisika T, Kazembe PN, Feluzi H, et al. Sexually Transmitted Infection (STI) screening, case and contact treatment, and condom promotion resulting in STI reduction two years later in rural Malawi. Malawi Med J. 2012;24:8–13.
CAS PubMed PubMed Central Google Scholar
Francis SC, Mthiyane TN, Baisley K, Mchunu SL, Ferguson JB, Smit T, et al. Prevalence of sexually transmitted infections among young people in South Africa: A nested survey in a health and demographic surveillance site. PLoS Med. 2018;15:e1002512.
de Lima YA, Turchi MD, Fonseca ZC, Garcia FL, de Brito E Cardoso FA, da Guarda Reis MN, et al. Sexually transmitted bacterial infections among young women in Central Western Brazil. Int J Infect Dis. 2014;25:16–21.
Carcamo CP, Campos PE, Garcia PJ, Hughes JP, Garnett GP, Holmes KK, et al. Prevalences of sexually transmitted infections in young adults and female sex workers in Peru: a national population-based survey. Lancet Infect Dis. 2012;12:765–73.
Sonnenberg P, Clifton S, Beddows S, Field N, Soldan K, Tanton C, et al. Prevalence, risk factors, and uptake of interventions for sexually transmitted infections in Britain: findings from the National Surveys of Sexual Attitudes and Lifestyles (Natsal). Lancet. 2013;382:1795–806.
Torrone EA, Johnson RE, Tian LH, Papp JR, Datta SD, Weinstock HS. Prevalence of Neisseria gonorrhoeae among persons 14 to 39 years of age, United States, 1999 to 2008. Sex Transm Dis. 2013;40:202–5.
Huai P, Li F, Li Z, Sun L, Fu X, Pan Q, et al. Prevalence, risk factors, and medical costs of Chlamydia trachomatis infections in Shandong Province, China: a population-based, cross-sectional study. BMC Infect Dis. 2018;18:534.
Luo ZZ, Li W, Wu QH, Zhang L, Tian LS, Liu LL, et al. Population-based study of chlamydial and gonococcal infections among women in Shenzhen. China: Implications for programme planning. PLoS One. 2018;13:e0196516.
Corsenac P, Noel M, Rouchon B, Hoy D, Roth A. Prevalence and sociodemographic risk factors of chlamydia, gonorrhoea and syphilis: a national multicentre STI survey in New Caledonia, 2012. BMJ Open. 2015;5:e007691.
Mulu W, Yimer M, Zenebe Y, Abera B. Common causes of vaginal infections and antibiotic susceptibility of aerobic bacterial isolates in women of reproductive age attending at Felegehiwot Referral Hospital. Ethiopia: a cross sectional study. BMC Womens Health. 2015;15:42.
Tadesse E, Teshome M, Amsalu A, Shimelis T. Genital Chlamydia trachomatis infection among women of reproductive age attending the gynecology clinic of Hawassa University Referral Hospital, Southern Ethiopia. PLoS One. 2016;11:e0168580.
Yirenya-Tawiah D, Annang TN, Apea-Kubi KA, Lomo G, Mensah D, Akyeh L, et al. Chlamydia Trachomatis and Neisseria Gonorrhoeae prevalence among women of reproductive age living in urogenital schistosomiasis endemic area in Ghana. BMC Res Notes. 2014;7:349.
Jespers V, Crucitti T, Menten J, Verhelst R, Mwaura M, Mandaliya K, et al. Prevalence and correlates of bacterial vaginosis in different sub-populations of women in sub-Saharan Africa: a cross-sectional study. PLoS One. 2014;9:e109670.
Kerubo E, Laserson KF, Otecko N, Odhiambo C, Mason L, Nyothach E, et al. Prevalence of reproductive tract infections and the predictive value of girls’symptom-based reporting: findings from a cross-sectional survey in rural western Kenya. Sex Transm Infect. 2016;92:251–6.
Masese LN, Wanje G, Kabare E, Budambula V, Mutuku F, Omoni G, et al. Screening for sexually transmitted infections in adolescent girls and young women in Mombasa, Kenya: feasibility, prevalence, and correlates. Sex Transm Dis. 2017;44:725–31.
Otieno FO, Ndivo R, Oswago S, Pals S, Chen R, Thomas T, et al. Correlates of prevalent sexually transmitted infections among participants screened for an HIV incidence cohort study in Kisumu, Kenya. Int J STD AIDS. 2015;26:225–37.
Menendez C, Castellsague X, Renom M, Sacarlal J, Quinto L, Lloveras B, et al. Prevalence and risk factors of sexually transmitted infections and cervical neoplasia in women from a rural area of southern Mozambique. Infect Dis Obstet Gynecol. 2010;2010:609315.
Kaida A, Dietrich JJ, Laher F, Beksinska M, Jaggernath M, Bardsley M, et al. A high burden of asymptomatic genital tract infections undermines the syndromic management approach among adolescents and young adults in South Africa: implications for HIV prevention efforts. BMC Infect Dis. 2018;18:499.
Peters RPH, Dubbink JH, van der Eem L, Verweij SP, Bos MLA, Ouburg S, et al. Cross-sectional study of genital, rectal, and pharyngeal chlamydia and gonorrhea in women in rural South Africa. Sex Transm Dis. 2014;41:564–9.
Rassjo EB, Mirembe F, Darj E. Self-reported sexual behaviour among adolescent girls in Uganda: reliability of data debated. Afr Health Sci. 2011;11:383–9.
Rutherford GW, Anglemyer A, Bagenda D, Muyonga M, Lindan CP, Barker JL, et al. University students and the risk of HIV and other sexually transmitted infections in Uganda: the Crane survey. Int J Adolesc Med Health. 2014;26:209–15.
Offorjebe OA, Wynn A, Moshashane N, Joseph Davey D, Arena K, Ramogola-Masire D, et al. Partner notification and treatment for sexually transmitted infections among pregnant women in Gaborone, Botswana. Int J STD AIDS. 2017;28:1184–9.
Wynn A, Ramogola-Masire D, Gaolebale P, Moshashane N, Agatha Offorjebe O, Arena K, et al. Acceptability and feasibility of sexually transmitted infection testing and treatment among pregnant women in Gaborone, Botswana, 2015. Biomed Res Int. 2016;2016:1251238.
Masha SC, Wahome E, Vaneechoutte M, Cools P, Crucitti T, Sanders EJ. High prevalence of curable sexually transmitted infections among pregnant women in a rural county hospital in Kilifi, Kenya. PLoS One. 2017;12:e0175166.
Warr AJ, Pintye J, Kinuthia J, Drake AL, Unger JA, McClelland RS, et al. Sexually transmitted infections during pregnancy and subsequent risk of stillbirth and infant mortality in Kenya: a prospective study. Sex Transm Infect. 2019;95:60–6.
Abdelaziz ZA, Ibrahim ME, Bilal NE, Hamid ME. Vaginal infections among pregnant women at Omdurman Maternity Hospital in Khartoum, Sudan. J Infect Dev Ctries. 2014;8:490–7.
Abdelrahim NA, Ahmed HI, Fadl-Elmula IM, Bayoumi MA, Homeida MM. Sexually transmitted infections other than HIV/AIDS among women of low socio-economic class attending antenatal clinics in Khartoum, Sudan. Int J STD AIDS. 2017;28:781–7.
Chiduo M, Theilgaard ZP, Bakari V, Mtatifikolo F, Bygbjerg I, Flanholc L, et al. Prevalence of sexually transmitted infections among women attending antenatal clinics in Tanga, north eastern Tanzania. Int J STD AIDS. 2012;23:325–9.
Hokororo A, Kihunrwa A, Hoekstra P, Kalluvya SE, Changalucha JM, Fitzgerald DW, et al. High prevalence of sexually transmitted infections in pregnant adolescent girls in Tanzania: a multi-community cross-sectional study. Sex Transm Infect. 2015;91:473–8.
Chaponda EB, Chico RM, Bruce J, Michelo C, Vwalika B, Mharakurwa S, et al. Malarial infection and curable sexually transmitted and reproductive tract infections among pregnant women in a rural district of Zambia. Am J Trop Med Hyg. 2016;95:1069–76.
Piazzetta RC, de Carvalho NS, de Andrade RP, Piazzetta G, Piazzetta SR, Carneiro R. Prevalence of Chlamydia trachomatis and Neisseria gonorrhoea infections in sexual actives young women at a southern Brazilian city. Rev Bras Ginecol Obstet. 2011;33:328–33.
PubMed Google Scholar
Pinto VM, Szwarcwald CL, Baroni C, Stringari LL, Inocencio LA, Miranda AE. Chlamydia trachomatis prevalence and risk behaviors in parturient women aged 15 to 24 in Brazil. Sex Transm Dis. 2011;38:957–61.
Rocha DA, Filho RA, Marino JM, dos Santos CM. “Hidden” sexually transmitted infections among women in primary care health services, Amazonas, Brazil. Int J STD AIDS. 2014;25:878–86.
Conejero C, Cannoni G, Merino PM, Bollmann J, Hidalgo C, Castro M, et al. Screening of Neisseria gonorrhoeae and Chlamydia trachomatis using techniques of self collected vaginal sample in young women. Rev Chil Infectol. 2013;30:489–93.
Article Google Scholar
Huneeus A, Schilling A, Fernandez MI. Prevalence of Chlamydia trachomatis, Neisseria gonorrhoeae, and Trichomonas vaginalis infection in Chilean adolescents and young adults. J Pediatr Adolesc Gynecol. 2018;31:411–5.
Paredes MC, Gomez YM, Torres AM, Fernandez M, Tovar MB. Prevalence of infections by Chlamydia trachomatis and Neisseria gonorrhoeae among high school students in the Sabana Central area of Cundinamarca, Colombia. Biomedica. 2015;35:314–24.
Jobe KA, Downey RF, Hammar D, Van Slyke L, Schmidt TA. Epidemiology of sexually transmitted infections in rural southwestern Haiti: the Grand’nse Women’s Health Study. Am J Trop Med Hyg. 2014;91:881–6.
Casillas-Vega N, Morfin-Otero R, Garcia S, Llaca-Diaz J, Rodriguez-Noriega E, Camacho-Ortiz A, et al. Sexually transmitted pathogens, coinfections and risk factors in patients attending obstetrics and gynecology clinics in Jalisco, Mexico. Salud Publica Mex. 2016;58:437–45.
Silveira MF, Erbelding EJ, Ghanem KG, Johnson HL, Burke AE, Zenilman JM. Risk of Chlamydia trachomatis infection during pregnancy: effectiveness of guidelines-based screening in identifying cases. Int J STD AIDS. 2010;21:367–70.
Bristow CC, Mathelier P, Ocheretina O, Benoit D, Pape JW, Wynn A, et al. Chlamydia trachomatis, Neisseria gonorrhoeae, and Trichomonas vaginalis screening and treatment of pregnant women in Port-au-Prince, Haiti. Int J STD AIDS. 2017;28:1130–4.
Pourabbas B, Rezaei Z, Mardaneh J, Shahian M, Alborzi A. Prevalence of Chlamydia trachomatis and Neisseria gonorrhoeae infections among pregnant women and eye colonization of their neonates at birth time, Shiraz, Southern Iran. BMC Infect Dis. 2018;18:477.
Hassan SJ, Dunphy E, Navin E, Marron L, Fitzsimmons C, Loy A, et al. Screening for chlamydia is acceptable and feasible during cervical screening in general practice. Ir Med J. 2016;109:326–7.
CAS PubMed Google Scholar
Matteelli A, Capelli M, Sulis G, Toninelli G, Carvalho AC, Pecorelli S, et al. Prevalence of Chlamydia trachomatis and Neisseria gonorrhoeae infection in adolescents in Northern Italy: an observational school-based study. BMC Public Health. 2016;16:200.
Salfa MC, Suligoi B. Prevalence of Chlamydia trachomatis, Trichomonas vaginalis and Neisseria gonorrhoeae based on data collected by a network of clinical microbiology laboratories, in Italy. Adv Exp Med Biol. 2016;901:47–57.
Nolskog P, Backhaus E, Nasic S, Enroth H. STI with Mycoplasma genitalium-more common than Chlamydia trachomatis in patients attending youth clinics in Sweden. Eur J Clin Microbiol Infect Dis. 2019;38:81–6.
Sakem B, Michel R, Nydegger UE, Radjenovic D, Wydler M, Risch M, et al. Diagnostic relevance of simultaneous testing for Chlamydia trachomatis and Neisseria gonorrhoeae. Infection. 2011;39:231–7.
Grech P, Marchant R, Samuel M. Sexual health of women aged 40 and over attending an inner city integrated sexual health clinic. Int J STD AIDS. 2017;28:404–7.
Peuchant O, Le Roy C, Desveaux C, Paris A, Asselineau J, Maldonado C, et al. Screening for Chlamydia trachomatis, Neisseria gonorrhoeae, and Mycoplasma genitalium should it be integrated into routine pregnancy care in French young pregnant women? Diagn Microbiol Infect Dis. 2015;82:14–9.
Borges-Costa J, Matos C, Pereira F. Sexually transmitted infections in pregnant adolescents: prevalence and association with maternal and foetal morbidity. J Eur Acad Dermatol Venereol. 2012;26:972–5.
Centers for Disease Control and Prevention (CDC). Sexually transmitted disease surveillance 2018: STDs in adolescents and young adults. https://www.cdc.gov/std/stats18/adolescents.htm . Accessed 9 Oct 2020.
Jackson JA, McNair TS, Coleman JS. Over-screening for chlamydia and gonorrhea among urban women age >/=25 years. Am J Obstet Gynecol. 2015;212:40.e1–6.
Newbern EC, Anschuetz GL, Eberhart MG, Salmon ME, Brady KA, De Los RA, et al. Adolescent sexually transmitted infections and risk for subsequent HIV. Am J Public Health. 2013;103:1874–81.
Nsuami MJ, Taylor SN. Most adolescents who participate in school-based screenings for sexually transmitted infections do not perceive themselves at high risk of sexually transmitted infection. Int J STD AIDS. 2012;23:822–4.
Akoh CC, Pressman EK, Cooper E, Queenan RA, Pillittere J, O’Brien KO. Prevalence and risk factors for infections in a pregnant adolescent population. J Pediatr Adolesc Gynecol. 2017;30:71–5.
Berggren EK, Patchen L. Prevalence of Chlamydia trachomatis and Neisseria gonorrhoeae and repeat infection among pregnant urban adolescents. Sex Transm Dis. 2011;38:172–4.
Blatt AJ, Lieberman JM, Hoover DR, Kaufman HW. Chlamydial and gonococcal testing during pregnancy in the United States. Am J Obstet Gynecol. 2012;207:55 e1–8.
Waight MT, Rahman MM, Soto P, Tran T. Sexually transmitted diseases during pregnancy in Louisiana, 2007-2009: high-risk populations and adverse newborn outcomes. J La State Med Soc. 2013;165:219–26.
Krishnan A, Sabeena S, Bhat PV, Kamath V, Hindol M, Zadeh VR, et al. Detection of genital chlamydial and gonococcal infection using urine samples: A community-based study from India. J Infect Public Health. 2018;11:75–9.
Asavapiriyanont S, Chaovarindr U, Kaoien S, Chotigeat U, Kovavisarach E. Prevalence of sexually transmitted infection in teenage pregnancy in Rajavithi Hospital, Thailand. J Med Assoc Thail. 2016;99(Suppl 2):S153–60.
Google Scholar
Choe HS, Lee DS, Lee SJ, Lee CB, Lee WC, Cho YH. Prevalence of sexually transmitted infections and sexual behavior of young adults and middle-aged people presenting to health examination centers in Korea. J Infect Chemother. 2012;18:207–12.
Kim Y, Kim J, Lee KA. Prevalence of sexually transmitted infections among healthy Korean women: implications of multiplex PCR pathogen detection on antibiotic therapy. J Infect Chemother. 2014;20:74–6.
Vallely LM, Toliman P, Ryan C, Rai G, Wapling J, Gabuzzi J, et al. Performance of syndromic management for the detection and treatment of genital Chlamydia trachomatis, Neisseria gonorrhoeae and Trichomonas vaginalis among women attending antenatal, well woman and sexual health clinics in Papua New Guinea: a cross-sectional study. BMJ Open. 2017;7:e018630.
Marks M, Kako H, Butcher R, Lauri B, Puiahi E, Pitakaka R, et al. Prevalence of sexually transmitted infections in female clinic attendees in Honiara, Solomon Islands. BMJ Open. 2015;5:e007276.
Ekeroma AJ, Pandit L, Bartley C, Ikenasio-Thorpe B, Thompson JMD. Screening for sexually transmitted infections in pregnancy at Middlemore Hospital, 2009. N Z Med J. 2012;125:23–9.
Badman SG, Vallely LM, Toliman P, Kariwiga G, Lote B, Pomat W, et al. A novel point-of-care testing strategy for sexually transmitted infections among pregnant women in high-burden settings: results of a feasibility study in Papua New Guinea. BMC Infect Dis. 2016;16:250.
Wangnapi RA, Soso S, Unger HW, Sawera C, Ome M, Umbers AJ, et al. Prevalence and risk factors for Chlamydia trachomatis, Neisseria gonorrhoeae and Trichomonas vaginalis infection in pregnant women in Papua New Guinea. Sex Transm Infect. 2015;91:194–200.
Downey RF, Hammar D, Jobe KA, Schmidt TA, Slyke LV, Yassemi Y, et al. Epidemiology of sexually transmitted infections in rural Haitian men. Int J STD AIDS. 2015;26:710–5.
Drinkard LN, Huxta RA, Halbritter A, Nguyen GT, Malebranche D. The case for extragenital screening of Chlamydia trachomatis and Neisseria gonorrhoeae in the college health setting. Sex Transm Dis. 2017;44:274–7.
Dave SS, Copas A, Richens J, White RG, Kosambiya JK, Desai VK, et al. HIV and STI prevalence and determinants among male migrant workers in India. PLoS One. 2012;7:e43576.
Jatapai A, Sirivongrangson P, Lokpichat S, Chuenchitra T, Nelson KE, Rangsin R. Prevalence and risk factors for Chlamydia trachomatis infection among young Thai men in 2008-2009. Sex Transm Dis. 2013;40:241–6.
Zhang G, Wong M, Yi P, Xu J, Li B, Ding G, et al. HIV-1 and STIs prevalence and risk factors of miners in mining districts of Yunnan, China. J Acquir Immune Defic Syndr. 2010;53(Suppl 1):S54–60.
European Centre for Disease Prevention and Control. Surveillance Atlas of Infectious Diseases. http://atlas.ecdc.europa.eu/public/index.aspx . Accessed 23 Nov 2020.
European Centre for Disease Prevention and Control (ECDC). Gonorrhoea. Annual Epidemiological Report for 2017. https://www.ecdc.europa.eu/sites/portal/files/documents/gonorrhoea-annual-epidemiological-report-2017.pdf . Accessed 23 Nov 2020.
Centers for Disease Control and Prevention (CDC). Sexually transmitted disease surveillance 2018: National profile - overview, Gonorrhea. https://www.cdc.gov/std/stats18/gonorrhea.htm . Accessed 9 Oct 2020.
Government of Canada - Public Health Agency of Canada. Notifiable disease charts. https://diseases.canada.ca/notifiable/charts-list . Accessed 30 Nov 2020.
World Health Organization. Report on global sexually transmitted infection surveillance 2015. http://apps.who.int/iris/bitstream/handle/10665/249553/9789241565301-eng.pdf;jsessionid=C5961C831C5BA711926D76292A64BAC2?sequence=1 . Accessed 30 Nov 2020.
Australasian Sexual Health Alliance. STI management guidelines for use in primary care. http://www.sti.guidelines.org.au/sexually-transmissible-infections/gonorrhoea . Accessed 30 Nov 2020.
Ministry of Health Singapore. Blood-borne and sexually transmitted diseases. https://www.moh.gov.sg/docs/librariesprovider5/resources-statistics/reports/blood-borne-and-sexually-transmitted-diseases.pdf . Accessed 30 Nov 2020.
Gonorrhoea writing group on behalf of the New Zealand Sexual Health Society. New Zealand guideline for the management of gonorrhoea, 2014, and response to the threat of antimicrobial resistance. https://nzshs.org/docman/guidelines/best-practice-guidelines/142-new-zealand-guideline-for-the-management-of-gonorrhoea-2014-and-response-to-the-threat-of-antimicrobial-resistance/file . Accessed 30 Nov 2020.
National Institute of Infectious Diseases. NESID Annual Surveillance Data Sentinel-Reporting Diseaes 2015-3. https://www.niid.go.jp/niid/en/survei/2085-idwr/ydata/6552-report-eb2015-3.html . Accessed 30 Nov 2020.
Korea Disease Control and Prevention Agency. Sexually Transmitted Infections (STIs) surveillance in the Republic of Korea, 2014-2018 - Public Health Weekly Report. http://www.cdc.go.kr/board/board.es?mid=a30501000000&bid=0031&list_no=366797&act=view . Accessed 17 Mar 2021.
Tafuma TA, Merrigan MB, Okui LA, Lebelonyane R, Bolebantswe J, Mine M, et al. HIV/sexually transmitted infection prevalence and sexual behavior of men who have sex with men in 3 districts of Botswana: results from the 2012 biobehavioral survey. Sex Transm Dis. 2014;41:480–5.
Sanders EJ, Wahome E, Okuku HS, Thiong’o AN, Smith AD, Duncan S, et al. Evaluation of WHO screening algorithm for the presumptive treatment of asymptomatic rectal gonorrhoea and chlamydia infections in at-risk MSM in Kenya. Sex Transm Infect. 2014;90:94–9.
Sanders EJ, Okuku HS, Smith AD, Mwangome M, Wahome E, Fegan G, et al. High HIV-1 incidence, correlates of HIV-1 acquisition, and high viral loads following seroconversion among MSM. AIDS. 2013;27:437–46.
Keshinro B, Crowell TA, Nowak RG, Adebajo S, Peel S, Gaydos CA, et al. High prevalence of HIV, chlamydia and gonorrhoea among men who have sex with men and transgender women attending trusted community centres in Abuja and Lagos, Nigeria. J Int AIDS Soc. 2016;19:21270.
Wade AS, Larmarange J, Diop AK, Diop O, Gueye K, Marra A, et al. Reduction in risk-taking behaviors among MSM in Senegal between 2004 and 2007 and prevalence of HIV and other STIs. ELIHoS Project, ANRS 12139. AIDS Care. 2010;22:409–14.
Rebe K, Lewis D, Myer L, de Swardt G, Struthers H, Kamkuemah M, et al. A cross sectional analysis of gonococcal and chlamydial infections among men-who-have-sex-with-men in Cape Town, South Africa. PLoS One. 2015;10:e0138315.
Ross MW, Nyoni J, Ahaneku HO, Mbwambo J, McClelland RS, McCurdy SA. High HIV seroprevalence, rectal STIs and risky sexual behaviour in men who have sex with men in Dar es Salaam and Tanga, Tanzania. BMJ Open. 2014;4:e006175.
Kim EJ, Hladik W, Barker J, Lubwama G, Sendagala S, Ssenkusu JM, et al. Sexually transmitted infections associated with alcohol use and HIV infection among men who have sex with men in Kampala, Uganda. Sex Transm Infect. 2016;92:240–5.
Cunha CB, Friedman RK, de Boni RB, Gaydos C, Guimaraes MRC, Siqueira BH, et al. Chlamydia trachomatis, Neisseria gonorrhoeae and syphilis among men who have sex with men in Brazil. BMC Public Health. 2015;15:686.
Creswell J, Guardado ME, Lee J, Nieto AI, Kim AA, Monterroso E, et al. HIV and STI control in El Salvador: results from an integrated behavioural survey among men who have sex with men. Sex Transm Infect. 2012;88:633–8.
Figueroa JP, Weir SS, Jones-Cooper C, Byfield L, Hobbs MM, McKnight I, et al. High HIV prevalence among men who have sex with men in Jamaica is associated with social vulnerability and other sexually transmitted infections. West Indian Med J. 2013;62:286–91.
Allan-Blitz LT, Leon SR, Bristow CC, Konda KA, Vargas SK, Flores JA, et al. High prevalence of extra-genital chlamydial or gonococcal infections among men who have sex with men and transgender women in Lima, Peru. Int J STD AIDS. 2017;28:138–44.
Castillo R, Konda KA, Leon SR, Silva-Santisteban A, Salazar X, Klausner JD, et al. HIV and sexually transmitted infection incidence and associated risk factors among high-risk MSM and male-to-female transgender women in Lima, Peru. J Acquir Immune Defic Syndr. 2015;69:567–75.
Kojima N, Park H, Konda KA, Joseph Davey DL, Bristow CC, Brown B, et al. The PICASSO Cohort: baseline characteristics of a cohort of men who have sex with men and male-to-female transgender women at high risk for syphilis infection in Lima, Peru. BMC Infect Dis. 2017;17:255.
Leon SR, Segura ER, Konda KA, Flores JA, Silva-Santisteban A, Galea JT, et al. High prevalence of Chlamydia trachomatis and Neisseria gonorrhoeae infections in anal and pharyngeal sites among a community-based sample of men who have sex with men and transgender women in Lima, Peru. BMJ Open. 2016;6:e008245.
Perez-Brumer AG, Konda KA, Salvatierra HJ, Segura ER, Hall ER, Montano SM, et al. Prevalence of HIV, STIs, and risk behaviors in a cross-sectional community- and clinic-based sample of men who have sex with men (MSM) in Lima, Peru. PLoS One. 2013;8:e59072.
Dudareva-Vizule S, Haar K, Sailer A, Wisplinghoff H, Wisplinghoff F, Marcus U, et al. Prevalence of pharyngeal and rectal Chlamydia trachomatis and Neisseria gonorrhoeae infections among men who have sex with men in Germany. Sex Transm Infect. 2014;90:46–51.
Marcus U, Ort J, Grenz M, Eckstein K, Wirtz K, Wille A. Risk factors for HIV and STI diagnosis in a community-based HIV/STI testing and counselling site for men having sex with men (MSM) in a large German city in 2011-2012. BMC Infect Dis. 2015;15:14.
Foschi C, Gaspari V, Sgubbi P, Salvo M, D’Antuono A, Marangoni A. Sexually transmitted rectal infections in a cohort of ‘men having sex with men’. J Med Microbiol. 2018;67:1050–7.
Heiligenberg M, Wermeling PR, van Rooijen MS, Urbanus AT, Speksnijder AGCL, Heijman T, et al. Recreational drug use during sex and sexually transmitted infections among clients of a city sexually transmitted infections clinic in Amsterdam, the Netherlands. Sex Transm Dis. 2012;39:518–27.
van Liere GAFS, Hoebe CJPA, Dukers-Muijrers NHTM. Evaluation of the anatomical site distribution of chlamydia and gonorrhoea in men who have sex with men and in high-risk women by routine testing: cross-sectional study revealing missed opportunities for treatment strategies. Sex Transm Infect. 2014;90:58–60.
van Liere GAFS, van Rooijen MS, Hoebe CJPA, Heijman T, de Vries HJ, Dukers-Muijrers NHTM. Prevalence of and factors associated with rectal-only chlamydia and gonorrhoea in women and in men who have sex with men. PLoS One. 2015;10:e0140297.
Soni S, Alexander S, Verlander N, Saunders P, Richardson D, Fisher M, et al. The prevalence of urethral and rectal Mycoplasma genitalium and its associations in men who have sex with men attending a genitourinary medicine clinic. Sex Transm Infect. 2010;86:21–4.
Gratrix J, Singh AE, Bergman J, Egan C, McGinnis J, Drews SJ, et al. Prevalence and characteristics of rectal chlamydia and gonorrhea cases among men who have sex with men after the introduction of nucleic acid amplification test screening at 2 Canadian sexually transmitted infection clinics. Sex Transm Dis. 2014;41:589–91.
Remis RS, Liu J, Loutfy MR, Tharao W, Rebbapragada A, Huibner S, et al. Prevalence of sexually transmitted viral and bacterial infections in HIV-positive and HIV-negative men who have sex with men in Toronto. PLoS One. 2016;11:e0158090.
Anschuetz GL, Paulukonis E, Powers R, Asbel LE. Extragenital screening in men who have sex with men diagnoses more chlamydia and gonorrhea cases than urine testing alone. Sex Transm Dis. 2016;43:299–301.
Hassan A, Blumenthal JS, Dube MP, Ellorin E, Corado K, Moore DJ, et al. Effect of rectal douching/enema on rectal gonorrhoea and chlamydia among a cohort of men who have sex with men on HIV pre-exposure prophylaxis. Sex Transm Infect. 2018;94:508–14.
Mayer KH, Ducharme R, Zaller ND, Chan PA, Case P, Abbott D, et al. Unprotected sex, underestimated risk, undiagnosed HIV and sexually transmitted diseases among men who have sex with men accessing testing services in a New England bathhouse. J Acquir Immune Defic Syndr. 2012;59:194–8.
Montano MA, Dombrowski JC, Dasgupta S, Golden MR, Duerr A, Manhart LE, et al. Changes in sexual behavior and STI diagnoses among MSM initiating PrEP in a clinic setting. AIDS Behav. 2019;23:548–55.
Mustanski B, Feinstein BA, Madkins K, Sullivan P, Swann G. Prevalence and risk factors for rectal and urethral sexually transmitted infections from self-collected samples among young men who have sex with men participating in the Keep it up! 2.0 randomized controlled trial. Sex Transm Dis. 2017;44:483–8.
Patton ME, Kidd S, Llata E, Stenger M, Braxton J, Asbel L, et al. Extragenital gonorrhea and chlamydia testing and infection among men who have sex with men--STD Surveillance Network, United States, 2010-2012. Clin Infect Dis. 2014;58:1564–70.
Sexton ME, Baker JJ, Nakagawa K, Li Y, Perkins R, Slack RS, et al. How reliable is self-testing for gonorrhea and chlamydia among men who have sex with men? J Fam Pract. 2013;62:70–8.
Taylor MM, Newman DR, Gonzalez J, Skinner J, Khurana R, Mickey T. HIV status and viral loads among men testing positive for rectal gonorrhoea and chlamydia, Maricopa County, Arizona, USA, 2011-2013. HIV Med. 2015;16:249–54.
Aggarwal P, Bhattar S, Sahani SK, Bhalla P, Garg VK. Sexually transmitted infections and HIV in self reporting men who have sex with men: A two-year study from India. J Infect Public Health. 2016;9:564–70.
Hananta IP, van Dam AP, Bruisten SM, van der Loeff MFS, Soebono H, de Vries HJ. Gonorrhea in Indonesia: high prevalence of asymptomatic urogenital gonorrhea but no circulating extended spectrum cephalosporins-resistant Neisseria gonorrhoeae strains in Jakarta, Yogyakarta, and Denpasar, Indonesia. Sex Transm Dis. 2016;43:608–16.
Morineau G, Nugrahini N, Riono P, Nurhayati GP, Mustikawati DE, et al. Sexual risk taking, STI and HIV prevalence among men who have sex with men in six Indonesian cities. AIDS Behav. 2011;15:1033–44.
Pattanasin S, Dunne EF, Wasinrapee P, Tongtoyai J, Chonwattana W, Sriporn A, et al. Screening for Chlamydia trachomatis and Neisseria gonorrhoeae infection among asymptomatic men who have sex with men in Bangkok, Thailand. Int J STD AIDS. 2018;29:577–87.
Tongtoyai J, Todd CS, Chonwattana W, Pattanasin S, Chaikummao S, Varangrat A, et al. Prevalence and correlates of Chlamydia trachomatis and Neisseria gonorrhoeae by anatomic site among urban Thai men who have sex with men. Sex Transm Dis. 2015;42:440–9.
Chow EPF, Tomnay J, Fehler G, Whiley D, Read TR, Denham I, et al. Substantial increases in chlamydia and gonorrhea positivity unexplained by changes in individual-level sexual behaviors among men who have sex with men in an Australian sexual health service from 2007 to 2013. Sex Transm Dis. 2015;42:81–7.
Nash JL, Hocking JS, Read TR, Chen MY, Bradshaw CS, Forcey DS, et al. Contribution of sexual practices (other than anal sex) to bacterial sexually transmitted infection transmission in men who have sex with men: a cross-sectional analysis using electronic health records. Sex Transm Infect. 2014;90:55–7.
Ong JJ, Fethers K, Howden BP, Fairley CK, Chow EPF, Williamson DA, et al. Asymptomatic and symptomatic urethral gonorrhoea in men who have sex with men attending a sexual health service. Clin Microbiol Infect. 2017;23:555–9.
Ryder N, Lockart IG, Bourne C. Is screening asymptomatic men who have sex with men for urethral gonorrhoea worthwhile? Sex Health. 2010;7:90–1.
Vodstrcil LA, Fairley CK, Fehler G, Leslie D, Walker J, Bradshaw CS, et al. Trends in chlamydia and gonorrhea positivity among heterosexual men and men who have sex with men attending a large urban sexual health service in Australia, 2002-2009. BMC Infect Dis. 2011;11:158.
Yang TZT, Chen MY, Read TRH, Needleman R, Bradshaw CS, Fortune R, et al. Sampling technique and detection rates of oropharyngeal and anorectal gonorrhoea using nucleic acid amplification tests in men who have sex with men. Sex Transm Infect. 2018;94:287–92.
Chen X, Fu G, Xu X, Hu H, Zuo H, Liu X, et al. Study on gonococcal and chlamydial infections among men who have sex with men in Nanjin. Acta Universitatis Medicinalis Anhui. 2011:569–72.
Fu GF, Jiang N, Hu HY, Mahapatra T, Yin YP, Mahapatra S, et al. The epidemic of HIV, syphilis, chlamydia and gonorrhea and the correlates of sexual transmitted infections among men who have sex with men in Jiangsu, China, 2009. PLoS One. 2015;10:e0118863.
Guo Y, Wang D, Zhou J, Chen S, Wang J, Zhen S, et al. Effects of education level of men who have sex with men on their high risk sexual behaviors and the infection of HIV and syphilis. Zhonghua Yu Fang Yi Xue Za Zhi. 2014;48:307–11.
Huan XP, Yin YP, Fu GF, Jiang N, Zhang QQ, Zhang XN, et al. Analysis on sexually transmitted diseases and the related risk factors among men who have sex with men in Jiangsu province. Zhonghua Yu Fang Yi Xue Za Zhi. 2011;45:975–8.
Liu YJ, Jiang SL, Hu Y, Song L, Yu M, Li SM. Characteristics of sexual behaviors and infection status of AIDS and other sexually transmitted diseases among men who have sex with men in 2009 in Beijing. Zhonghua Yu Fang Yi Xue Za Zhi. 2011;45:971–4.
Yang LG, Zhang XH, Zhao PZ, Chen ZY, Ke WJ, Ren XQ, et al. Gonorrhea and chlamydia prevalence in different anatomical sites among men who have sex with men: a cross-sectional study in Guangzhou. China. BMC Infect Dis. 2018;18:675.
Zhang X, Jia M, Chen M, Luo H, Chen H, Luo W, et al. Prevalence and the associated risk factors of HIV, STIs and HBV among men who have sex with men in Kunming, China. Int J STD AIDS. 2017;28:1115–23.
Jung M, Lee J, Kwon DS, Park BJ. Comparison of sexual risky factors of men who have sex with men and sex-buying men as groups vulnerable to sexually transmitted diseases. J Prev Med Public Health. 2012;45:156–63.
Pham QD, Nguyen TV, Hoang CQ, Cao V, Khuu NV, Phan HT, et al. Prevalence of HIV/STIs and associated factors among men who have sex with men in An Giang, Vietnam. Sex Transm Dis. 2012;39:799–806.
Beymer MR, Weiss RE, Bolan RK, Rudy ET, Bourque LB, Rodriguez JP, et al. Sex on demand: geosocial networking phone apps and risk of sexually transmitted infections among a cross-sectional sample of men who have sex with men in Los Angeles County. Sex Transm Infect. 2014;90:567–72.
Cheung KT, Fairley CK, Read TRH, Denham I, Fehler G, Bradshaw CS, et al. HIV incidence and predictors of incident HIV among men who have sex with men attending a sexual health clinic in Melbourne, Australia. PLoS One. 2016;11:e0156160.
Pinsky L, Chiarilli DB, Klausner JD, Kull RM, O’Keefe R, Heffer C, et al. Rates of asymptomatic nonurethral gonorrhea and chlamydia in a population of university men who have sex with men. J Am Coll Heal. 2012;60:481–4.
Behanzin L, Diabate S, Minani I, Boily MC, Labbe AC, Ahoussinou C, et al. Decline in the prevalence of HIV and sexually transmitted infections among female sex workers in Benin over 15 years of targeted interventions. J Acquir Immune Defic Syndr. 2013;63:126–34.
Merrigan MB, Tafuma TA, Okui LA, Lebelonyane R, Bolebantswe JM, Makhaola K, et al. HIV prevalence and risk behaviors among female sex workers in Botswana: results from the 2012 HIV/STI bio-behavioral study. AIDS Behav. 2015;19:899–908.
Vuylsteke B, Semde G, Sika L, Crucitti T, Ettiegne Traore V, Buve A, et al. HIV and STI prevalence among female sex workers in Cote d’Ivoire: why targeted prevention programs should be continued and strengthened. PLoS One. 2012;7:e32627.
Tadele A, Hussen S, Shimelis T. Prevalence and associated factors of Chlamydia trachomatis and Neisseria gonorrhoeae among female commercial sex workers in Hawassa City, Southern Ethiopia. BMC Infect Dis. 2019;19:61.
Aho J, Koushik A, Coutlee F, Diakite SL, Rashed S. Prevalence of HIV, human papillomavirus type 16 and herpes simplex virus type 2 among female sex workers in Guinea and associated factors. Int J STD AIDS. 2014;25:280–8.
Izulla P, McKinnon LR, Munyao J, Karanja S, Koima W, Parmeres J, et al. HIV postexposure prophylaxis in an urban population of female sex workers in Nairobi, Kenya. J Acquir Immune Defic Syndr. 2013;62:220–5.
Braunstein SL, Ingabire CM, Kestelyn E, Uwizera AU, Mwamarangwe L, Ntirushwa J, et al. High human immunodeficiency virus incidence in a cohort of Rwandan female sex workers. Sex Transm Dis. 2011;38:385–94.
Vandepitte J, Muller E, Bukenya J, Nakubulwa S, Kyakuwa N, Buve A, et al. Prevalence and correlates of Mycoplasma genitalium infection among female sex workers in Kampala, Uganda. J Infect Dis. 2012;205:289–96.
Sabido M, Lahuerta M, Montoliu A, Gonzalez V, Hernandez G, Giardina F, et al. Human immunodeficiency virus, sexually transmitted infections, and risk behaviors among clients of sex workers in Guatemala: are they a bridge in human immunodeficiency virus transmission? Sex Transm Dis. 2011;38:735–42.
Tinajeros F, Miller WM, Castro L, Artiles N, Flores F, Evans JL, et al. Declining sexually transmitted infections among female sex workers: the results of an HIV and sexually transmitted infection prevention strategy in Honduras, 2006-08. Int J STD AIDS. 2012;23:88–93.
Bazzi AR, Rangel G, Martinez G, Ulibarri MD, Syvertsen JL, Bazzi SA, et al. Incidence and predictors of HIV and sexually transmitted infections among female sex workers and their intimate male partners in northern Mexico: a longitudinal, multilevel study. Am J Epidemiol. 2015;181:723–31.
Kazerooni PA, Motazedian N, Motamedifar M, Sayadi M, Sabet M, Lari MA, et al. The prevalence of human immunodeficiency virus and sexually transmitted infections among female sex workers in Shiraz, South of Iran: by respondent-driven sampling. Int J STD AIDS. 2014;25:155–61.
Nasirian M, Kianersi S, Hoseini SG, Kassaian N, Yaran M, Shoaei P, et al. Prevalence of sexually transmitted infections and their risk factors among female sex workers in Isfahan, Iran: a cross-sectional study. J Int Assoc Provid AIDS Care. 2017;16:608–14.
Khan MS, Unemo M, Zaman S, Lundborg CS. HIV, STI prevalence and risk behaviours among women selling sex in Lahore, Pakistan. BMC Infect Dis. 2011;11:119.
Znazen A, Frikha-Gargouri O, Berrajah L, Bellalouna S, Hakim H, Gueddana N, et al. Sexually transmitted infections among female sex workers in Tunisia: high prevalence of Chlamydia trachomatis. Sex Transm Infect. 2010;86:500–5.
Mc Grath-Lone L, Marsh K, Hughes G, Ward H. The sexual health of female sex workers compared with other women in England: analysis of cross-sectional data from genitourinary medicine clinics. Sex Transm Infect. 2014;90:344–50.
Haseen F, Chawdhury FA, Hossain ME, Huq M, Bhuiyan MU, Imam H, et al. Sexually transmitted infections and sexual behaviour among youth clients of hotel-based female sex workers in Dhaka, Bangladesh. Int J STD AIDS. 2012;23:553–9.
Khanam R, Reza M, Ahmed D, Rahman M, Alam MS, Sultana S, et al. Sexually transmitted infections and associated risk factors among street-based and residence-based female sex workers in Dhaka, Bangladesh. Sex Transm Dis. 2017;44:21–8.
Das A, Pathni AK, Narayanan P, George B, Morineau G, Saidel T, et al. High rates of reinfection and incidence of bacterial sexually transmitted infections in a cohort of female sex workers from two Indian cities: need for different STI control strategies? Sex Transm Infect. 2013;89:5–10.
Hemalatha R, Kumar RH, Venkaiah K, Srinivasan K, Brahmam GN. Prevalence of & knowledge, attitude & practices towards HIV & sexually transmitted infections (STIs) among female sex workers (FSWs) in Andhra Pradesh. Indian J Med Res. 2011;134:470–5.
Bollen LJ, Anartati AS, Morineau G, Sulami S, Prabawanti C, Silfanus FJ, et al. Addressing the high prevalence of gonorrhoea and chlamydia among female sex workers in Indonesia: results of an enhanced, comprehensive intervention. Sex Transm Infect. 2010;86:61–5.
Majid N, Bollen L, Morineau G, Daily SF, Mustikawati DE, Agus N, et al. Syphilis among female sex workers in Indonesia: need and opportunity for intervention. Sex Transm Infect. 2010;86:377–83.
Mawu FO, Davies SC, McKechnie M, Sedyaningsih ER, Widihastuti A, Hillman RJ. Sexually transmissible infections among female sex workers in Manado, Indonesia, using a multiplex polymerase chain reaction-based reverse line blot assay. Sex Health. 2011;8:52–60.
Silitonga N, Davies SC, Kaldor J, Wignall S, Okoseray M. Prevalence over time and risk factors for sexually transmissible infections among newly-arrived female sex workers in Timika, Indonesia. Sex Health. 2011;8:61–4.
Tanudyaya FK, Rahardjo E, Bollen LJ, Madjid N, Daili SF, Priohutomo S, et al. Prevalence of sexually transmitted infections and sexual risk behavior among female sex workers in nine provinces in Indonesia, 2005. Southeast Asian J Trop Med Public Health. 2010;41:463–73.
Couture MC, Sansothy N, Sapphon V, Phal S, Sichan K, Stein E, et al. Young women engaged in sex work in Phnom Penh, Cambodia, have high incidence of HIV and sexually transmitted infections, and amphetamine-type stimulant use: new challenges to HIV prevention and risk. Sex Transm Dis. 2011;38:33–9.
Chen XS, Yin YP, Liang GJ, Wang QQ, Jiang N, Liu Q, et al. The prevalences of Neisseria gonorrhoeae and Chlamydia trachomatis infections among female sex workers in China. BMC Public Health. 2013;13:121.
Guo Y, Xu X, Fu G, Huan X, Jiang N, Yin Y, et al. Risk behaviours and prevalences of HIV and sexually transmitted infections among female sex workers in various venues in Changzhou, China. Int J STD AIDS. 2017;28:1135–42.
Jin X, Chan S, Ding G, Wang H, Xu J, Wang G, et al. Prevalence and risk behaviours for Chlamydia trachomatis and Neisseria gonorrhoeae infection among female sex workers in an HIV/AIDS high-risk area. Int J STD AIDS. 2011;22:80–4.
Luo L, Li X, Zhang LL. Neisseria gonorrhoeae prevalence, incidence and associated risk factors among female sex workers in a high HIV-prevalence area of China. Int J Infect Dis. 2015;38:115–20.
Remis RS, Kang L, Calzavara L, Pan Q, Liu J, Myers T, et al. Prevalence and correlates of HIV infection and sexually transmitted infections in female sex workers (FSWs) in Shanghai, China. Epidemiol Infect. 2015;143:258–66.
Tang W, Pan J, Jiang N, Hu HY, Mahapatra T, Yin YP, et al. Correlates of chlamydia and gonorrhea infection among female sex workers: the untold story of Jiangsu, China. PLoS One. 2014;9:e85985.
Wong WC, Yim YL, Lynn H. Sexually transmitted infections among female sex workers in Hong Kong: the role of migration status. J Travel Med. 2011;18:1–7.
Wong HT, Lee KC, Chan DP. Community-based sexually transmitted infection screening and increased detection of pharyngeal and urogenital Chlamydia trachomatis and Neisseria gonorrhoeae infections in female sex workers in Hong Kong. Sex Transm Dis. 2015;42:185–91.
Zhu BY, Bu J, Huang PY, Zhou ZG, Yin YP, Chen XS, et al. Epidemiology of sexually transmitted infections, HIV, and related high-risk behaviors among female sex workers in Guangxi Autonomous Region, China. Jpn J Infect Dis. 2012;65:75–8.
Vuylsteke B, Semde G, Sika L, Crucitti T, Ettiegne Traore V, Buve A, et al. High prevalence of HIV and sexually transmitted infections among male sex workers in Abidjan, Cote d’Ivoire: need for services tailored to their needs. Sex Transm Infect. 2012;88:288–93.
Galarraga O, Sosa-Rubi SG, Gonzalez A, Badial-Hernandez F, Conde-Glez CJ, Juarez-Figueroa L, et al. The disproportionate burden of HIV and STIs among male sex workers in Mexico City and the rationale for economic incentives to reduce risks. J Int AIDS Soc. 2014;17:19218.
Goldsamt LA, Clatts MC, Giang LM, Le BQ, Colby DJ, Yu G. HIV and other STIs in male sex workers: Findings from a sexual health promotion intervention in Vietnam. Int J STD AIDS. 2018;29:540–6.
Chow EP, Tucker JD, Wong FY, Nehl EJ, Wang Y, Zhuang X, et al. Disparities and risks of sexually transmissible infections among men who have sex with men in China: a meta-analysis and data synthesis. PLoS One. 2014;9:e89959.
Institute of Health Metrics and Evaluation (IHME). Findings from the Global Burden of Disease Study 2017. http://www.healthdata.org/policy-report/findings-global-burden-disease-study-2017 . Accessed 30 Nov 2020.
World Health Organization. Standard protocol to assess prevalence of gonorrhoea and chlamydia among pregnant women in antenatal care clinics. https://apps.who.int/iris/handle/10665/275846 . Accessed 30 Nov 2020.
Vandenbroucke JP, von Elm E, Altman DG, Gøtzsche PC, Mulrow CD, Pocock SJ, et al. Strengthening the Reporting of Observational Studies in Epidemiology (STROBE): explanation and elaboration. PLoS Med. 2007;4:e297.
Download references
Acknowledgments
The authors thank the Modis platform for editorial assistance and manuscript coordination, on behalf of GSK. Kristel Vercauteren provided medical writing support and Maria Ana de la Grandière and Sara Blancquaert coordinated the manuscript development and provided editorial support. The authors also thank Masoud Dara and Giorgi Kuchukhidze (WHO European Regional Office, Copenhagen, Denmark); Jane Rowley (London, UK); and Nicholas J. Kassebaum (Institute for Health Metrics and Evaluation, IHME, Seattle, USA).
This work was funded by GlaxoSmithKline Biologicals SA. MU was not funded.
Author information
Authors and affiliations.
Clinical and Epidemiology Research and Development, GSK, Amsterdam, The Netherlands
Jane Whelan & Victoria Abbing-Karahagopian
Clinical and Epidemiology Research and Development, GSK, Siena, Italy
Laura Serino
WHO Collaborating Centre for Gonorrhoea and other STIs, Department of Laboratory Medicine, Microbiology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
Magnus Unemo
You can also search for this author in PubMed Google Scholar
Contributions
JW supervised this work. JW, VAK, LS and MU contributed to the conceptualization, investigation, methodology, validation, and visualization of this work. JW, VAK, LS and MU participated in the development and the review of the manuscript and approved the final submitted version.
Corresponding authors
Correspondence to Jane Whelan or Victoria Abbing-Karahagopian .
Ethics declarations
Ethics approval and consent to participate.
Not applicable.
Consent for publication
Competing interests.
VAK, LS and JW are employed by the GSK group of companies. JW and LS hold shares as part of their remuneration. MU declares no financial interests. None of the authors declare non-financial interests.
Additional information
Publisher’s note.
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Additional file 1..
PRISMA checklist.
Additional file 2.
Literature search strategy.
Additional file3.
Inclusion and exclusion criteria.
Additional file 4.
Standardizations adopted for urogenital, rectal and pharyngeal laboratory tests.
Additional file 5.
Reported gonorrhoea prevalence and/or test positivity in men-who-have-sex-with-men, by WHO region, country, and anatomic site.
Additional file 6.
Dataset supporting the output of the literature search.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ . The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Reprints and permissions
About this article
Cite this article.
Whelan, J., Abbing-Karahagopian, V., Serino, L. et al. Gonorrhoea: a systematic review of prevalence reporting globally. BMC Infect Dis 21 , 1152 (2021). https://doi.org/10.1186/s12879-021-06381-4
Download citation
Received : 07 April 2021
Accepted : 09 June 2021
Published : 11 November 2021
DOI : https://doi.org/10.1186/s12879-021-06381-4
Share this article
Anyone you share the following link with will be able to read this content:
Sorry, a shareable link is not currently available for this article.
Provided by the Springer Nature SharedIt content-sharing initiative
- Neisseria gonorrhoeae
- systematic review
- men-who-have-sex-with-men
- sex workers
BMC Infectious Diseases
ISSN: 1471-2334
- General enquiries: [email protected]
Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.
- View all journals
- Explore content
- About the journal
- Publish with us
- Sign up for alerts
- Published: 21 November 2019
- Magnus Unemo 1 , 2 ,
- H Steven Seifert 3 ,
- Edward W. Hook III 4 ,
- Sarah Hawkes 5 ,
- Francis Ndowa 6 &
- Jo-Anne R. Dillon 7 , 8
Nature Reviews Disease Primers volume 5 , Article number: 79 ( 2019 ) Cite this article
65k Accesses
293 Citations
159 Altmetric
Metrics details
- Antimicrobial resistance
- Bacterial infection
- Infectious-disease diagnostics
- Microbial genetics
- Urogenital diseases
The bacterium Neisseria gonorrhoeae causes the sexually transmitted infection (STI) gonorrhoea, which has an estimated global annual incidence of 86.9 million adults. Gonorrhoea can present as urethritis in men, cervicitis or urethritis in women, and in extragenital sites (pharynx, rectum, conjunctiva and, rarely, systemically) in both sexes. Confirmation of diagnosis requires microscopy of Gram-stained samples, bacterial culture or nucleic acid amplification tests. As no gonococcal vaccine is available, prevention relies on promoting safe sexual behaviours and reducing STI-associated stigma, which hinders timely diagnosis and treatment thereby increasing transmission. Single-dose systemic therapy (usually injectable ceftriaxone plus oral azithromycin) is the recommended first-line treatment. However, a major public health concern globally is that N. gonorrhoeae is evolving high levels of antimicrobial resistance (AMR), which threatens the effectiveness of the available gonorrhoea treatments. Improved global surveillance of the emergence, evolution, fitness, and geographical and temporal spread of AMR in N. gonorrhoeae , and improved understanding of the pharmacokinetics and pharmacodynamics for current and future antimicrobials in the treatment of urogenital and extragenital gonorrhoea, are essential to inform treatment guidelines. Key priorities for gonorrhoea control include strengthening prevention, early diagnosis, and treatment of patients and their partners; decreasing stigma; expanding surveillance of AMR and treatment failures; and promoting responsible antimicrobial use and stewardship. To achieve these goals, the development of rapid and affordable point-of-care diagnostic tests that can simultaneously detect AMR, novel therapeutic antimicrobials and gonococcal vaccine(s) in particular is crucial.
Similar content being viewed by others
Antimicrobial treatment and resistance in sexually transmitted bacterial infections
Multidrug-resistant Neisseria gonorrhoeae infection in heterosexual men with reduced susceptibility to ceftriaxone, first report in Thailand
Investigation of auranofin and gold-containing analogues antibacterial activity against multidrug-resistant Neisseria gonorrhoeae
Introduction.
The sexually transmitted infection (STI) gonorrhoea remains a major public health concern globally. The aetiological agent of gonorrhoea, the bacterium Neisseria gonorrhoeae (the gonococcus), generally causes mucosal infections of the urogenital tract, predominantly infecting columnar and transitional epithelia, although it can also attach to the stratified squamous epithelium of the ectocervix 1 , 2 . Such N. gonorrhoeae infections most frequently result in urethritis in men and cervicitis in women, but urethritis in women is also observed 3 , 4 . This obligate human host-adapted pathogen was described for the first time by Albert Neisser in Gram-stained microscopy of urethral discharge in 1879 (ref. 5 ). N. gonorrhoeae is a diplococcal (that is, it is typically composed of two joined cells with the adjacent sides flattened, resulting in a characteristic kidney or coffee bean appearance on microscopy), Gram-negative microorganism; it belongs to the bacterial class Betaproteobacteria and the family Neisseriaceae, and has been co-evolving with its human host for centuries. The family Neisseriaceae comprises the genus Neisseria and other genera such as Kingella and Eikenella 6 , 7 , 8 . The Neisseria genus currently consists of at least 23 species, of which about half are human-restricted species, some are animal-restricted and some can be isolated from mucosal surfaces in both humans and animals 8 . N. gonorrhoeae is genomically, morphologically and phenotypically closely related to the other pathogenic Neisseria species, Neisseria meningitidis , which is typically carried as a commensal in the (naso)pharynx of 10–15% of the general population but occasionally causes fatal septicaemia and/or meningitis 6 , 8 , 9 , 10 . N. gonorrhoeae is also related to several other commensal Neisseria species that reside particularly in the pharynx. Despite containing many of the pathogenicity and virulence factors of N. gonorrhoeae and N. meningitidis , the commensal Neisseria species, from which these two pathogenic Neisseria species have evolved, do not normally cause pathology 9 as they are unable to induce substantial polymorphonuclear leukocyte (PMNL)-based inflammation and lack several additional factors and mechanisms of interacting with host molecules, cells and tissues 11 . The pathogenesis and pathophysiology of N. gonorrhoeae have been studied for decades; however, detailed knowledge regarding many fundamental properties is lacking.
The majority of men with gonococcal urethritis are symptomatic, but substantially fewer women with urogenital gonorrhoea are symptomatic and, when present, symptoms are nonspecific. Nevertheless, signs of infection can be identified in most women with urogenital gonorrhoea. Rectal and pharyngeal gonorrhoea, which is mostly asymptomatic, are most frequently diagnosed in men who have sex with men (MSM), but are not rare in women either. Disseminated gonococcal infections (DGIs) are rare but can occur in both adults and neonates 6 , 12 , 13 . If infections are not detected and/or adequately treated, ascending infections, such as epididymitis and salpingitis, can result in a variety of serious complications and sequelae, particularly in women who bear the major burden of disease; these complications and sequelae include pelvic inflammatory disease (PID), chronic pelvic pain, ectopic pregnancy and infertility. Gonorrhoea also facilitates the transmission and acquisition of other STIs including HIV infection. Gonococcal infections can lead to complications during pregnancy, and infected women can also transmit infections to children during birth causing ophthalmia neonatorum, which was a leading cause of blindness in the pre-antimicrobial era. Conjunctivitis in adults is also observed sporadically. Thus, gonorrhoea causes substantial morbidity and socioeconomic consequences globally 12 , 14 , 15 .
In the absence of a gonococcal vaccine, management and control rely on effective, affordable and accessible antimicrobial treatment, supported by adequate prevention, diagnostic testing or screening, notification and management of sex partners of infected individuals, and epidemiological surveillance. However, N. gonorrhoeae has developed or acquired antimicrobial resistance (AMR) to all antimicrobials recommended earlier as first-line or second-line empirical treatment of gonorrhoea (for example, sulfonamides, penicillins, tetracyclines, fluoroquinolones, early-generation macrolides, such as erythromycin, and cephalosporins, such as cefuroxime). This extensive resistance has been accomplished by an accumulation of AMR determinants, most of which do not seem to substantially reduce the biological fitness of the bacterium 16 , 17 , 18 , 19 , 20 , 21 (Fig. 1 ). This AMR is of serious public health concern as the pathogen has become highly resistant to all previously recommended antimicrobials, and resistance to the currently recommended extended-spectrum cephalosporin (ESC) ceftriaxone and macrolide azithromycin has also emerged. On the basis of the high global prevalence of gonorrhoea, the high level of antimicrobial use and/or misuse, suboptimal diagnosis, limited control and surveillance of AMR, suboptimal or slow update of management guidelines, and the extraordinary ability of N. gonorrhoeae to acquire or develop — and retain — AMR, it is likely that the global impact of gonorrhoea, including its severe complications and sequelae, will increase, and further N. gonorrhoeae AMR will evolve in the future. Consequently, improved global actions and research efforts to retain gonorrhoea as a readily treatable infection are essential.
Each bar represents a gonorrhoea therapy, and the length of the bar represents the time period from when the therapy started to be used until when clinical and/or in vitro resistance threatening the effectiveness of that specific antimicrobial therapy emerged. In vitro verified antimicrobial resistance (AMR) determinants are also shown 16 , 17 , 18 , 19 , 20 , 21 , 218 , 219 , 220 . PBP2 amino acid alterations that increase the minimum inhibitory concentration (MIC) of extended-spectrum cephalosporins (ESCs; verified, for example, by site-directed mutagenesis or transformation) in nonmosaic and mosaic (in which concomitant epistatic mosaic penA mutations are also needed) penA alleles are noted by an asterisk 218 , 219 , 220 . Additionally, PBP2 G542S, P551S, and P551L amino acid alterations in nonmosaic penA alleles have been statistically associated with gonococcal strains with decreased susceptibility to ESCs 221 , 222 , 223 . A grave concern is that during the past decade(s) resistance to azithromycin and decreased susceptibility to the ESC ceftriaxone, the last remaining option for empirical monotherapy, have been reported worldwide. The first Neisseria gonorrhoeae strain with high-level resistance to ceftriaxone was isolated in 2009 in Japan, which was followed by some isolates with high-level ceftriaxone resistance in 2011 in France and Spain. During subsequent years, ceftriaxone-resistant isolates have been characterized in many countries including Japan, China, Australia, Singapore, Canada, Argentina and several European countries. Furthermore, treatment failures with ceftriaxone were verified in Japan, Australia and in several European countries 15 , 16 , 153 , 224 , 225 , 226 , 227 , 228 , 229 , 230 , 231 , 232 , 233 , 234 , 235 , 236 , 237 , 238 , 239 , 240 . In 2014, the first failure of ceftriaxone–azithromycin dual therapy for gonorrhoea was verified in the UK 241 . Worryingly, since 2015, an international spread of one ceftriaxone-resistant gonococcal strain, initially described in Japan, has been confirmed 229 , 230 , 231 , 232 , 233 , 234 , 235 , 239 , 240 , 242 , 243 , and the first strain with resistance to ceftriaxone plus high-level azithromycin resistance was isolated in 2018 in the UK and Australia 236 , 237 , 238 . rRNA, ribosomal RNA; SNP, single-nucleotide polymorphism.
This Primer focuses on the epidemiology, aetiological agent, pathogenic mechanisms/pathophysiology, diagnosis, screening, prevention and management of gonorrhoea. We also discuss global actions and research efforts imperative for future management and control of gonorrhoea.
Epidemiology
In 2016, the WHO estimated that there were 86.9 (95% uncertainty interval 58.6–123.4) million incident global cases of gonorrhoea (global prevalence 0.9%) among adults 15–49 years of age 22 (Fig. 2 ). The epidemiological diversity of gonorrhoea manifests itself in the variability of the geographical distribution and the prevalence among certain populations; determinants of such variability include sexuality and sexual orientation, socioeconomics, demographics, geographical and cultural ramifications (including stigma and taboos), and access to and quality of sex education, prevention, testing and diagnostics, as well as political commitment in the provision of health services 23 , 24 , 25 .
Estimated numbers (in millions) of incident cases of gonorrhoea in adults (15–49 years of age) by WHO region 22 . These data correspond to 20 new gonococcal infections per 1,000 women and 26 per 1,000 men globally. The highest incidence was in the WHO African region, with 41 cases per 1,000 women and 50 per 1,000 men, followed by the WHO region of the Americas, with 23 cases per 1,000 women and 32 per 1,000 men; the lowest incidence was in the WHO European region, with 7 cases per 1,000 women and 11 per 1,000 men 22 . The World Bank Income Classification is also shown. Data from ref. 22 .
Epidemiological determinants
When individual countries, especially in industrialized settings, embarked on prevention and care of STIs on the basis of the established determinants of STIs, declines in rates of gonococcal infections were observed during the late 1980s. However, this decline was short-lived, as increases in gonococcal infections rates have been reported since the late 1990s. Observations have identified a number of factors, both established and new, as important to explain the high rates of STIs, including gonococcal infections; these factors include ethnic background, sexuality and sexual preferences, sexual mixing patterns, such as assortative mixing by race and/or ethnicity (that is, the tendency to connect with individuals of the same race and/or ethnicity) and disassortative mixing by risk group (that is, the tendency to connect with individuals with a different risk level), gender and disparities in economic status and access to services, as well as the intrinsic characteristics of the pathogen 24 , 26 , 27 , 28 , 29 , 30 .
Other reasons for the recent increase in gonorrhoea incidence in many high-resourced settings include changes in sexual behaviour in the era of antiretroviral treatment for HIV infection (that is, because of the availability of antiretroviral treatment and the perception that HIV infection is no longer life-threatening in the short term, people are less cautious and have sex with new and casual partners without using condoms), increased electronic connectivity (for example, the use of dating apps for meeting sex partners), increased number of casual unknown partners, larger sexual networks, increased travel and variable access to services 30 , 31 . Another factor to be taken into consideration is the increasing use of drugs in sexual networks, particularly common among MSM and female sex workers. Finally, certain key populations are at higher risk of and disproportionately affected by STIs, including gonorrhoea; such populations include MSM, migrants, young people and sex workers.
Incidence and prevalence
The aforementioned factors, mostly in combination, probably substantially contributed to the varying increases in gonorrhoea case rates in the past 5–10 years, even in countries with more comprehensive health systems. For example, in the USA and in the European Union/European Economic Area (EU/EEA), both socioeconomic status and ethnic background have been observed to highly correlate with gonococcal infection rates. In the USA in 2017, the rate of reported cases of gonorrhoea was approximately eight times higher among black populations than among white populations. Higher rates were also noted among American Indians and Alaskan Natives, Native Hawaiians and individuals with Hispanic heritage, whereas the rate among individuals with Asian heritage was half the rate among white individuals 30 , 31 . In the USA, the number of gonorrhoea cases increased by 67% from 2013 ( n = 333,510) to 2017 ( n = 556,413) 32 . The proportion of gonococcal isolates cultured from MSM increased from 3.9% in 1989 to a high of 38.5% in 2017, reflecting epidemiological changes and possibly changes in the health care-seeking behaviour of men with gonorrhoea as well as improved reporting of sexual orientation in the USA 30 , 31 .
In the EU/EEA, the number of reported gonorrhoea cases has increased by >200% since 2008, from 29,434 cases in 2008 (with an incidence of 7.85 per 100,000 population) to 89,239 cases in 2017, with the highest numbers of cases in the UK, France, the Netherlands and Spain 33 . Of note, higher prevalence in these countries might be in part accounted for by the availability of comprehensive sexual health systems, frequent testing and/or surveillance. The highest incidence of gonorrhoea in the EU/EEA is in young adults (15–24 years of age) 33 . MSM accounted for ~25–30% of all the cases in the EU/EEA during recent years — 30% of the reported gonorrhoea cases (57% of the cases reporting sexual orientation) in Europe in 2017 (ref. 33 ); however, over the past decade, substantial increases also occurred among heterosexual men, men with no sexual orientation reported and women. In the UK, MSM experienced substantial increases in reported STIs in 2017. Of the 50,032 new nonviral STI diagnoses in MSM in 2017, 43% were gonococcal infections and, between 2016 and 2017, gonococcal infection diagnoses increased by 21% 34 .
The geographical setting in which people live also seems to have a role in the prevalence of gonococcal infection, probably reflecting differences in the access to information regarding STIs, availability, accessibility and quality of health-care services, and social factors such as the effect of stigma on health-care-seeking behaviours. Observations showed that the prevalence of gonorrhoea in women aged 15–24 years in clinical or community settings in South Africa was ~4.6%, whereas in southern Africa and eastern Africa the prevalence was 1.7%. Furthermore, in the same study, the prevalence in a high-risk population in eastern Africa, mostly sex workers, was 8.2% 35 .
In low-income settings, syndromic management of STIs is mainly performed, and no comprehensive aetiology-based surveillance systems enable accurate assessment nationwide of increases or decreases in gonorrhoea prevalence in the general population or in subpopulations. However, even in many high-income settings, for example in Europe, the surveillance data should be interpreted with caution as the surveillance systems, testing, methodologies and quality assurance are not standardized across countries and remain weak in several settings 33 , 36 . Finally, whole-genome sequencing (WGS) will revolutionize our understanding of the epidemiology of gonorrhoea and the geographical and temporal spread of AMR and antimicrobial susceptible N. gonorrhoeae strains in different populations and subpopulations, including at-risk groups (see Outlook, below).
Gonorrhoea in MSM on pre-exposure prophylaxis
Another topical area of interest is the observation of rapid increases in the incidence of gonorrhoea, and other STIs, in high-resourced settings among MSM taking pre-exposure prophylaxis (PrEP) for the prevention of HIV infection. Some published data reported that MSM using PrEP can be ~25 times more likely to acquire a gonococcal infection than MSM not using PrEP 37 . A multisite open-label study of just under 3,000 gay and bisexual men using PrEP, conducted in Australia between 2016 and 2018, showed a significant increase in the incidence of STIs (including gonorrhoea, Chlamydia trachomatis infection and syphilis) during a follow-up period of 1.1 years. Younger age, greater number of sex partners and group sex participation were associated with a greater risk for an STI, whereas inconsistent or no condom use with casual partners was not 38 . A systematic review commissioned by the WHO in 2018–2019 identified 88 STI studies, primarily in MSM in high-income countries, which found that STI prevalence was high in people prior to starting PrEP, and STI incidence varied by setting and population included in the review. However, pooled STI incidence generally remained high during follow-up when taking PrEP 39 , 40 . However, notably, individuals on PrEP are monitored more closely and tested more frequently for STIs than non-PrEP users. When both populations were controlled for frequent monitoring, as in the PROUD study, no statistically significant differences in STI rates were found between men taking PrEP and the control group 41 . Thus, it would seem that the reduced risk for and fear of HIV infection has led some PrEP users, especially young MSM, to reduce condom use and/or increase other risky sexual behaviours and, therefore, to place themselves at increased exposure to other STIs, including gonorrhoea. However, given the conflicting conclusions from different population studies on this point, more observations and studies are needed to identify the factors behind these contradictory conclusions, as well as to detail the risk factors and elements that may be responsible for the findings of increased STI risk in some populations and to better understand the ideal monitoring and screening intervals of individuals taking PrEP.
Mechanisms/pathophysiology
The bacterium N. gonorrhoeae
Growth and metabolism.
N. gonorrhoeae is a fastidious organism that is sensitive to many environmental factors such as oxygen, nonphysiological temperatures, desiccation and the presence of toxic substances (such as many fatty acids), among others 42 ; thus, the bacterium does not survive for long outside the human host, and is difficult to culture (Box 1 ). Many strains have incomplete biosynthetic capabilities for amino acids, presumably because amino acids and other important nutrients are readily obtained from the human host. Iron (which is essential for bacterial growth) is acquired from the host by binding iron-containing host proteins such as transferrin, lactoferrin and haemoglobin at the bacterial surface and stripping these molecules of iron that is then delivered to the bacterial cytoplasm 43 . Owing to the broad range of oxygen levels within different niches of the male and female urogenital tracts, it is possible that N. gonorrhoeae encounters aerobic, microaerobic, and anaerobic conditions within the host, and the bacteria are able to grow in all these conditions 44 .
Box 1 Models to study Neisseria gonorrhoeae pathogenesis
Much of the information concerning Neisseria gonorrhoeae pathogenesis has come from studying the physiological and genetic properties of the organism, including determination of growth and nutrient requirements and surface-exposed molecules, with in vitro bacterial cultures. However, these experimental conditions do not always mirror in vivo conditions and, therefore, cell culture models can be useful to learn about the interactions between the bacterium and the host, particularly how N. gonorrhoeae attaches to and is internalized into eukaryotic cells. These studies have mainly used immortalized transformed human cell lines, but have occasionally used newly harvested human primary cells 1 , as cell lines do not always replicate the properties of tissues. Primary cultures are difficult to isolate and maintain and are considerably heterogeneous, whereas tissue explants enable the study of the interactions of the organism with different cell types in a complex tissue. Compared with other primary tissues, fallopian tube tissue is relatively easy to obtain from hysterectomies and is a clinically relevant tissue environment 70 , particularly for modelling pelvic inflammatory disease 212 .
Animal models are useful to study colonization, growth and immune response in a host. Of note, because N. gonorrhoeae is restricted to the human host, the bacterial proteins have evolved highly specific interactions with human molecules, rendering early mouse models of limited value. Despite this limitation, female mice treated with 17β-oestradiol (to promote prolonged colonization and/or infection) have become a standard in the field 193 . Transgenic mouse models expressing human receptors for N. gonorrhoeae are in development and will have greater utility in the future 194 , 195 , although no existing mouse model totally mimics a natural human infection. In the 1960s, primate models were examined and chimpanzees reportedly developed symptomatic gonorrhoea 213 , but chimpanzees are no longer used for biomedical research in the USA and rarely elsewhere, although new primate models might be developed in the future. The human challenge model is the most relevant existing model 214 . Only men can participate, as they have a lower risk of complications from infection than women. This model has only been used to investigate initial colonization determinants, and its utility is limited owing to small cohorts per study, the requirement for treatment as soon as symptoms develop and being only applicable to men.
Using WGS, it has been shown that the modern gonococcal population is not as old as previously thought and has been shaped by antimicrobial treatment of STIs as well as other infections, leading to the emergence of two major genomic lineages, one multidrug-resistant and one multidrug-susceptible, with different evolutionary strategies 45 . N. gonorrhoeae has a single circular chromosome between ~2.1 and 2.3 megabase pairs ( ∼ 2,200–2,500 protein-coding sequences), which exists as diploid, homozygous, chromosomes 46 , 47 . In addition, N. gonorrhoeae can acquire additional DNA via horizontal genetic transfer (HGT), the noninherited external acquisition of new genetic material from another bacterium. HGT occurs mainly by type IV pilus-mediated DNA transformation (uptake of DNA from the environment and subsequent incorporation into the genome). N. gonorrhoeae is naturally competent for transformation during its entire life cycle, but transformation only occurs at high frequency between cells of N. gonorrhoeae and other Neisseria species. Approximately 80% of isolates carry a chromosomal insertion called the gonococcal genetic island, which has genes similar to those carried on the conjugal plasmid (that is, genes involved in conjugation — the DNA transfer between bacteria by cell-to-cell contact). However, in N. gonorrhoeae these conjugation gene products act to secrete chromosomal DNA into the medium that is then available for DNA transformation. Pilus-mediated DNA transformation provides efficient transport of DNA into the bacterial cell and DNA uptake sequences are highly represented in Neisseria genomes ( ∼ 1,900–2,000 copies per genome) 48 , 49 . This efficient transformation is one reason why AMR determinants efficiently spread from cell to cell. Notably, this ability of N. gonorrhoeae to transfer DNA between strains makes clonal analysis difficult because alleles are not stably linked and led to the creation of the multilocus sequence typing system to characterize bacterial lineages by the DNA sequence type of several defined and more conserved housekeeping genes 50 . Multilocus sequence typing systems are now available for many different bacterial species 51 . Furthermore, this reassortment of alleles suggests that mixed-strain gonorrhoea infections are common 52 , 53 , although widely unrecognized, as most clinical laboratories analyse and save single colonies when culturing isolates, probably underestimating the incidence of mixed infections. Ideally, multiple colonies should be tested.
Nearly all gonococcal strains contain a cryptic plasmid (with no defined functions); many contain a plasmid encoding a penicillinase (mostly TEM-1 or TEM-135 β-lactamase), which results in high-level penicillin resistance, and conjugative plasmids, which sometimes carry tetM causing high-level tetracycline resistance, although these plasmids are not as prevalent as reported for many other bacterial species 16 , 54 . Several penicillinase-encoding plasmids of different size have been described in N. gonorrhoeae and named according to their epidemiological origin, such as the widely spread and most common African, Asian and Rio/Toronto plasmids. Different conjugative gonococcal plasmids carrying tetM have also been described, the most common being the American tetM plasmid and the Dutch tetM plasmid 16 , 54 . In addition, several double-stranded and single-stranded bacteriophage gene islands have been annotated within the N. gonorrhoeae genome, but no isolated bacteriophage that can infect and lyse the bacteria has been found 55 .
Colonization determinants
N. gonorrhoeae shares many colonization determinants with other human-restricted Neisseria species that rarely cause infection. The factors required to establish a host niche include the type IV pilus, the opacity protein family (Opa proteins), the porin PorB, efflux pumps and metal transport systems (Fig. 3 ). N. gonorrhoeae probably has to compete with the resident microbiota for colonization, but little is known about how different resident commensal organisms may limit or cooperate with N. gonorrhoeae during colonization.
Neisseria gonorrhoeae is a Gram-negative bacterium, frequently encountered as diplococci (individual cells are ∼ 0.6–1 µm in diameter), with a characteristic cell envelope consisting of a cytoplasma membrane (the inner membrane), a periplasmic space containing the peptidoglycan cell wall 244 and the outer membrane containing lipo-oligosaccharide (LOS), which is similar to lipopolysaccharide (LPS) of other Gram-negative bacteria, except it does not have the polymeric O-antigen characteristic of LPS. The type IV pilus is a long, thin fibre that reaches far outside of the cell envelope, mainly composed of many copies of one protein, pilin. Type IV pilus assembly requires a complex molecular machine, called the assembly apparatus, that sits within the cell envelope to produce the fibre on the outside of the cell 245 . The pilus is a dynamic structure that can be retracted by the assembly apparatus, which generates one of the largest physical forces on record by a biological machine 246 . The Opa proteins are a family of integral outer membrane proteins whose expression is stochastically controlled 247 . Each N. gonorrhoeae isolate carries ~11 opa genes, and expression of each is controlled by independent molecular events that turn on or off the expression of each opa gene. A single bacterial cell may express none of the Opa proteins, a single Opa, or a combination of several. There is a correlation between patterns of Opa expression and bacteria isolated from females during menses 248 , and increased numbers of Opa proteins are expressed during human volunteer infections 249 . The outer-membrane-localized porin (PorB) allows small molecules to enter the periplasm and the reduction modifiable protein (Rmp) is associated with PorB and elicits antibodies that block the binding of anti-PorB antibodies 250 . The five efflux pump systems (FarA–FarB–MtrE, NorM, MtrC–MtrD–MtrE, MacA–MacB–MtrE and MtrF) have varying substrate specificity and many roles in pathogenesis, including removing toxic molecules encountered during infection and exporting antimicrobials (acting as resistance determinants). The three iron-scavenging complexes (LpbA–LpbB, HpuA–HpuB and TbpA–TbpB) are required to obtain iron from the host. Adapted from ref. 5 , Springer Nature Limited.
Gonococcal pili are required for efficient mucosal colonization (typically of nonciliated columnar epithelia) and carry out many functions, including initial adherence to host cells and tissues, self-adherence and adherence to other N. gonorrhoeae cells, a means to crawl along mucosal surfaces called twitching motility, protection from PMNL killing mechanisms 56 , and HGT by DNA transformation 57 . Clinical isolates of N. gonorrhoeae are always piliated, but quickly lose pilus expression in laboratory culture through a variety of mechanisms, showing that pilus expression is under strong selective pressure during infection.
The Opa proteins mainly act as adhesins that bind to a variety of receptors found on many different cells and tissues 58 and mediate more intimate attachment and initiation of microcolony formation. Most Opa proteins bind to one or more human carcinoembryonic antigen-related cell adhesion molecules (CEACAMs), a family of surface-exposed proteins. Opa proteins only bind to human forms of these proteins, and a few Opa proteins also bind to heparan sulfate proteoglycans. Although some Opa–CEACAM interactions lead to cell signalling events, such as induction of the oxidative burst from PMNLs, most Opa interactions seem to be important for adherence to cells and tissues 59 .
All Gram-negative bacterial porins (transmembrane channel proteins) act to allow small molecules access to the periplasm. The N. gonorrhoeae porin (PorB) is one of the most abundant proteins in the outer membrane; it increases attachment, is then translocated to the host cell mitochondria and impairs the ability of phagocytes to kill the bacteria. Other important properties include resisting the action of complement factors, modulating apoptosis, invasion of host cells and involvement in AMR 60 , 61 , 62 , 63 .
N. gonorrhoeae expresses up to five efflux pump systems: MtrC–MtrD–MtrE, MacA–MacB–MtrE, NorM, FarA–FarB–MtrE and MtrF 64 , 65 , 66 . These export pumps have varying narrow or extensive substrate specificity and have many roles in pathogenesis, including removing toxic molecules encountered during infection, such as fatty acids and cationic peptides, and removing antimicrobials from the cell (that is, acting as AMR determinants). Finally, there are three iron acquisition systems in the envelope of N. gonorrhoeae , and each can strip iron from a human protein that is designed to sequester iron from pathogenic organisms. There is an acquisition system for transferrin (TbpA–TbpB), one for lactoferrin (LbpA–LbpB) and one for haeme (HpuA–HpuB), which can be found, for example, in haemoglobin 43 .
Infection dynamics
All bacteria that live in or on people need to colonize and grow, whether they are commensal organisms that rarely cause harm or frank pathogens. The pathogenesis field defines colonization and growth determinants as virulence determinants even though they are often found also within organisms that do not cause overt pathology. However, for a pathogenic organism to do damage, it usually needs to colonize specific anatomical sites and grow (except when pathogenesis occurs through production of a toxin away from the site of infection).
Transmission
N. gonorrhoeae infects the mucosal epithelium of the male and female urogenital tracts, the rectum, pharynx or conjunctiva 12 . N. gonorrhoeae is mainly transmitted through unprotected vaginal, anal or oral intercourse. During vaginal sex, transmission rates from men to women are higher than from women to men 67 . Ejaculate from infected men contains millions of bacteria, effectively injecting the organism into the receiving anatomical site. How the organism is effectively transmitted from vaginal, rectal or oral/pharyngeal locations to the male urethra is not completely understood. Of note, N. gonorrhoeae infection amplifies the risk for acquisition and transmission of HIV and several other STIs 68 , 69 ; all the underlying mechanisms are not completely understood, but probably involve factors such as inflammation, destruction of the mucosa and discharges. Furthermore, women with N. gonorrhoeae infection can effectively transmit the infection to their children during birth (intrapartum), but not during pregnancy; the neonate’s conjunctiva is highly exposed during transit of the birth canal, and N. gonorrhoeae infection of the conjunctiva results in ophthalmia neonatorum.
Host defences against infection act at many levels. N. gonorrhoeae has no ability to persist on or to penetrate the skin, and it requires a mucous membrane for colonization. Many barriers in mammalian cells limit transit of organisms into the body, including the ciliary action of some epithelia. Peptidoglycan fragments and lipo-oligosaccharide (LOS) released by N. gonorrhoeae can disrupt the ciliary action of the epithelium and may promote colonization 70 , 71 . Once colonization is established, innate and adaptive immune responses act to block or limit the growth of an organism. However, as a host-restricted organism that has co-evolved with its human host, N. gonorrhoeae has intricate mechanisms to limit the action of these host defence systems.
Innate immune systems
Resident tissue macrophages are one of the first cells that N. gonorrhoeae encounters during infection 72 (Fig. 4 ). Whether macrophages have a role in limiting N. gonorrhoeae infection is not clear, but macrophages, dendritic cells and epithelial cells may all be responsible for producing the chemokines and cytokines induced during infection. Some of these host effectors are responsible for inducing the massive PMNL response that manifests as the purulent exudate characteristic of symptomatic urethral gonorrhoea. N. gonorrhoeae can survive the various antimicrobial functions of PMNLs including phagocytosis, the release of reactive oxygen species, cationic peptides and antimicrobial enzymes, metal sequestration and PMNL extracellular traps 73 . N. gonorrhoeae can also modulate the apoptosis of epithelial cells, macrophages, T cells and PMNLs, but as both the inhibition and enhancement of apoptosis has been reported, the relevance of apoptosis modulation to infection remains controversial 74 , 75 . In addition, the role of PMNLs during N. gonorrhoeae infection also remains controversial. PMNLs probably influence infection by killing some of the bacteria but enabling the spread of others 73 .
Initial adhesion of Neisseria gonorrhoeae to the epithelium requires type IV pili and, then, Opa proteins for more intimate adhesion. The bacteria can then proliferate on the epithelial surface and invade underlying tissues via transcytosis. N. gonorrhoeae also releases peptidoglycan fragments, outer membrane vesicles (OMVs) and lipo-oligosaccharide (LOS), thereby activating Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain-containing protein signalling in tissue-resident dendritic cells and macrophages. In response to bacterial stimulation, these cells produce chemokines and cytokines (for example, IL-1, IL-6, IL-8, IL-17 and tumour necrosis factor (TNF)) that can recruit polymorphonuclear leukocytes (PMNLs); however, the bacteria can often survive phagocytosis, antibacterial factors released during degranulation or NETosis (that is, cell death mediated by neutrophil extracellular traps (NETs)). N. gonorrhoeae has many ways to prevent complement killing by the membrane attack complex; for example, the LOS can be modified by sialic acid, when the precursor substrate, CMP-NANA, is supplied by the host, to enhance complement resistance 251 . Sialylated LOS binds C3b and promotes its inactivation to iC3b via factor I, whereas the outer-membrane-localized porin (PorB) binds factor H and C4BP, thereby hiding the bacteria from complement recognition. When complement activity is inhibited (for example, by mutation or owing to immune suppressive treatment), systemic N. gonorrhoeae infections are prevalent. Whether the resistance to complement is also important in localized sites of colonization is not known. C4BP, C4b-binding protein; NLR, nucleotide-binding oligomerization domain-containing protein (NOD)-like receptor. Adapted from ref. 5 , Springer Nature Limited.
The classical and alternative complement pathways act to kill many organisms, and N. gonorrhoeae has evolved ways to avoid both pathways during uncomplicated infections 76 . Indicative of its extreme host restriction and evolution, N. gonorrhoeae remains sensitive to animal complement system components 61 . N. gonorrhoeae uses several mechanisms to limit complement-mediated killing by blocking deposition or activity of several complement factors 61 (Fig. 4 ). People with complement deficiencies are at increased risk of DGI, showing that the complement system helps to limit gonococcal survival in the blood stream 77 . Increased incidence of DGI and other disseminated Neisseria spp. infections was observed when patients were treated with eculizumab, a complement inhibitor, but this study did not report altered rates of uncomplicated gonorrhoea 78 . Whether complement effectively functions at mucosal sites of colonization is not fully known.
Adaptive immunity
As an organism that has co-evolved with its sole host for centuries, and possibly throughout all recorded time, the colonization determinants of N. gonorrhoeae are exquisitely adapted to life within humans. By contrast, the human adapted immune system has variable components (B cells and T cells) that can change to limit infection. N. gonorrhoeae is generally thought to be immunosuppressive 79 , although there are suggestions that any immunosuppression is incomplete. Many studies show that antigonococcal antibodies are found in people with active or previous infection, demonstrating a humoral immune response 80 . In addition, the existence of three, independent, antigenically variable surface antigens (type IV pilus, Opa proteins and LOS) also provides evidence that there are potentially protective responses directed against these antigens that necessitate the complex variations 81 . These antigens can all vary during infection and colonization; for example, the surface-exposed antigenic epitopes of pili will vary and pilus expression can be lost, the number and type of expressed Opa proteins will vary (Fig. 3 ), and the type of sugars on the LOS molecule can change. Although some of this surface variation alters some functional properties of N. gonorrhoeae , the most important function of antigenic variation is immune avoidance, which enables reinfection presumably even with the same gonococcal strain, as protective immunity to N. gonorrhoeae capable of preventing subsequent infections has never been recorded. Extensive surface molecule variation by N. gonorrhoeae also prevents these molecules from being considered viable vaccine candidates. A more detailed examination of immune suppression and responses during human infection is needed.
Host damage
N. gonorrhoeae is not a very disruptive pathogen, as it is well adapted to its human host and rarely lethal. It does not produce any exotoxins that can destroy host cells, but does secrete peptidoglycan fragments, outer membrane vesicles (OMVs) and LOS that are toxic to mammalian cells and can specifically inhibit the ciliated cells on fallopian tube tissues 70 , 71 . Moreover, when PMNLs are recruited to sites of infection, PMNL antimicrobial products are released that can damage the tissue. All of these factors contribute to the damage and scarring of the fallopian tube tissue that is characteristic of PID. These factors can also cause damage at other sites of infection, particularly during DGIs in which, in addition to fever, dermatitis, infectious arthritis and (less frequently) septicaemia, endocarditis and meningitis can occur.
Diagnosis, screening and prevention
Clinical presentation and diagnosis.
The incubation period for urogenital gonorrhoea ranges from ~2 days to 8 days 82 . The clinical manifestations of gonorrhoea are variable and differ markedly in men and women 12 . At least 90% of men with gonococcal urethritis are symptomatic, presenting with obvious urethral discharge and dysuria, a fact that permits the application of syndromic diagnosis (based on a set of symptoms and signs that are characteristic of a clinical manifestation) in many settings as both a time-saving and cost-saving measure. For men with symptomatic urethritis, Gram stain may be used to support symptom evaluation. By contrast, laboratory-based diagnostic tests have a more important role for gonococcal detection in asymptomatic men, women and in patients of all genders for extragenital (rectal and pharyngeal) infections, which are mostly asymptomatic or present with nonspecific symptoms. Although ~40% of women with gonococcal cervicitis may report abnormal vaginal discharge, this symptom is unreliable for syndromic diagnosis of gonorrhoea, as many other equally or more common genitourinary infections in women (for example, bacterial vaginosis, trichomoniasis and vaginal candidiasis) may cause the same symptoms.
Microbiological diagnosis of gonorrhoea can be challenging, as many regions do not have a laboratory-based diagnostic capability and rely on syndromic management algorithms to guide empirical antimicrobial treatments 14 . Microbiological diagnosis is performed by the detection of Gram-negative diplococci in stained smears using microscopy, culture of N. gonorrhoeae and/or nucleic acid amplification tests (NAATs) detecting N. gonorrhoeae DNA or RNA.
Traditional diagnostic methods
In resource-limited settings, light microscopy of Gram-stained samples is often the only method available to diagnose infection with N. gonorrhoeae presumptively (Table 1 ). The sensitivity and specificity of the Gram stain, which tests for the presence of characteristic Gram-negative diplococci within PMNLs, can vary substantially between studies and depends upon the specimen; the highest sensitivity and specificity were reported with urethral swab samples from symptomatic males (89% to >98% and >95%, respectively) 6 , 13 , 83 , 84 , 85 , whereas the sensitivity was as low as 40–50% in urethral specimens from asymptomatic males, and in endocervical or urethral specimens from women 13 , 83 , 84 . This difference can probably be explained by a reduced bacterial load, particularly in these urethral samples, and by the presence of many other bacterial species in the endocervical samples. Gram stain is not suitable for the diagnosis of N. gonorrhoeae from pharyngeal specimens (because other Neisseria species with similar morphology are prevalent in the oral and nasopharyngeal cavity) or rectal specimens (which have a sensitivity ≤40%) 82 , 83 , 84 . A methylene blue staining method is an alternative to the Gram stain, and similar high sensitivity and specificity were reported for diagnosing gonococcal urethritis in men 86 .
Prior to the introduction of NAATs, culture (Table 1 ) of the organism was the gold standard and this remains the only diagnostic method available in some settings as it is a low-cost method. Culture also remains recommended for test-of-cure for treatment failure, in cases of sexual abuse and to evaluate PID 13 , 85 , 87 . Furthermore, complete AMR testing can only be accomplished if N. gonorrhoeae is cultured 83 , 85 , 87 , 88 . Culture performance is dependent upon factors such as anatomical site of the cultured sample, method of specimen collection, media and conditions used to transport the sample to the diagnostic centre 83 , 87 , 89 , nonselective and/or selective culture media 84 , 85 , 89 , 90 , conditions of incubation 82 , 85 and species confirmatory tests. Cultures obtained too soon after exposure (<48 h) may give false-negative results 13 , and a repeated culture sample some weeks later is sometimes considered. Culture of urogenital specimens usually has a sensitivity ranging from 72% to 95%, but can have a sensitivity of 95–100% in settings with extensive experience in appropriate specimen handling and culture 83 , 84 . However, the sensitivity of culturing pharyngeal and rectal specimens is much lower.
Presumptive identification of cultured N. gonorrhoeae isolates is frequently accomplished by typical colony appearance on selective media, Gram-stained microscopy and the oxidase test, which detects the presence of cytochrome oxidase 82 , 84 , 85 . For definitive N. gonorrhoeae identification, immunological tests frequently targeting PorB 85 , 91 , 92 , 93 , sugar utilization tests or other biochemical tests 6 , 85 , 91 , 94 , NAATs or mass spectrometry (that is, matrix-associated laser desorption ionization time of flight (MALDI-TOF mass spectrometry)) 6 , 95 , 96 , 97 are frequently performed. These tests differentiate N. gonorrhoeae from species such as N. meningitidis , Neisseria lactamica , Neisseria cinerea, Neisseria subflava or other genera that may occasionally grow on even the selective culture media and may be present particularly in the pharynx but also at other sites 85 . Finally, DNA extraction from cultured isolates is also currently the best method to obtain DNA for genomic analysis, as clinical specimens often either do not contain sufficient concentrations of DNA, or contain too much DNA from other bacterial species or human cells. Furthermore, methods for genomic DNA purification from clinical specimens have not been sufficiently developed or standardized 98 .
NAATs are currently recommended for gonorrhoea diagnosis in most high-income countries 13 , 82 , 87 , 99 . NAATs are now the preferred diagnostic test because specimen collection is noninvasive (urine or self-collected particularly vaginal swabs); viable organisms are not required for detection, permitting less stringent transportation and storage methods 85 , 100 ; most have superior sensitivity with maintained high specificity (which vary between NAATs and anatomical site tested) compared with culture; they produce more rapid results (many later generation NAAT platforms allow for high throughput and automation); and many can simultaneously detect other STI-associated pathogens (particularly C. trachomatis ) 13 , 85 , 87 , 101 . Initially, a number of in-house, PCR-based NAATs were used locally and continue to be used as confirmatory tests or for diagnosis in resource-limited settings 93 , 102 , 103 , 104 . In-house NAATs generally target conserved regions of genes such as the porA pseudogene, opa genes, gyrA (encoding DNA gyrase subunit A) , cppB (encoding cryptic plasmid protein B) and the methyltransferase genes of N. gonorrhoeae 102 . Few reports have compared the performance of such in-house NAATs with culture or commercially available NAATs 102 . In high-income countries, in-house NAATs have largely been replaced with commercial NAATs that have been comprehensively validated and received regulatory approval from the US FDA 13 , 87 , 101 (Table 2 ).
In 2019, the first two NAATs (Aptima Combo 2 assay and Xpert CT/NG) for gonococcal detection received FDA approval also for extragenital specimens such as rectal and pharyngeal infection 105 , and licensing for additional NAATs is in progress. Several studies indicate that many additional NAATs are more sensitive, with maintained high specificity, than culture for diagnosing N. gonorrhoeae from pharyngeal and rectal specimens (Table 2 ); however, such tests should be used only after rigorous local performance evaluations 82 , 87 , 106 , and additionally a confirmatory NAAT with a different target should be used for such specimens 82 , 85 , 87 , 100 , as other Neisseria species, which can be frequently present especially in the pharynx, could be misidentified as N. gonorrhoeae 87 , 100 . Thus, when using NAATs to detect N. gonorrhoeae , it is important to choose the test or the testing strategy so that the positive predictive value (which is calculated based on the sensitivity and specificity of the test and on the local prevalence of the pathogen, and the last two parameters substantially affect the positive predictive value) is >90% 82 , 85 .
The introduction of NAATs for N. gonorrhoeae has substantially reduced the number of cultured patient samples. FDA-approved NAATs are more expensive than culture-based methods, and are mostly used in high-income countries 13 , 82 , 87 , 99 . Pooling specimens (that is, combining up to 5–10 specimens and then retesting them separately if the pool is positive to ascertain which specimen(s) was positive) may reduce cost, especially in settings with high-volume testing and with low positivity rate. However, strict evaluation of the performance characteristics of the NAAT in the local population is crucial before implementing any pooling strategy. Time to results, hands-on time, maintenance and consumption of reagents and consumables for automated platforms vary greatly between platforms, and these parameters influence the choice of platform 107 , 108 . A major disadvantage of commercial NAATs is the inability to perform AMR testing on gonococcal specimens 14 , 85 , 102 , 109 . In many regions, >80% of gonorrhoea cases are diagnosed by NAATs and, therefore, crucial information regarding AMR and gonococcal strain biology is lost. There are no recommended molecular tests for the prediction of antimicrobial susceptibility or resistance 102 , 110 , 111 ; however, a PCR-based test that also detects ciprofloxacin susceptibility status has received the European Conformity In Vitro Diagnostic mark (Table 3 ) and several NAATs in the pipeline are also being developed to detect both N. gonorrhoeae and its ciprofloxacin susceptibility status 101 . This type of test could be important particularly in regions in which ciprofloxacin susceptible strains are still spreading and, therefore, ciprofloxacin could be used for treatment as a lower cost, oral alternative to ceftriaxone plus azithromycin, that is to spare the use of these antimicrobials and decrease the selective pressure for resistance. This concept has been tested clinically with success 101 , 112 , 113 . Notably, both the British Association for Sexual Health and HIV (BASHH) gonorrhoea guideline for the UK and the European gonorrhoea guideline for the WHO European region recommend the use of ciprofloxacin for treatment of anogenital and pharyngeal gonorrhoea if the gonococcal strain causing the infection is proven to be ciprofloxacin-susceptible using genetic or phenotypic resistance testing 82 , 114 .
Point-of-care tests (POCTs)
Development of appropriate rapid point-of-care tests (POCTs) is a high priority for the diagnosis of gonorrhoea 14 , 85 , 101 , 115 (Table 3 ). POCTs could provide a definitive, rapid diagnosis to guide specific treatment in situations where this is not currently possible, such as in settings in which only syndromic management is available, in cases where patients may not return for treatment and for screening asymptomatic patients 116 , 117 , 118 . Ideally, POCTs should meet the ‘ASSURED’ criteria, that is, be affordable, sensitive, specific, user-friendly, robust and rapid, and equipment free (or requiring minimal equipment powered by solar or battery sources) 117 , 119 , 120 . However, all diagnostic tests that provide rapid test results and correct treatment during a single clinical visit could be defined as POCTs 117 , 121 , 122 . The Gram stain is an oft-used POCT; its benefits and limitations have been described above 122 , 123 . Other POCTs developed for N. gonorrhoeae include lateral flow immunochromatographic and optical immunoassay tests based on antigen detection, as well as a near-POCT NAAT — the Xpert CT/NG assay 101 , 120 , 122 , 123 . Recent reviews of the performance of several POCTs have shown that immunochromatographic-based and optical immunoassay-based POCTs had highly suboptimal sensitivities, some as low as 12.5%, and specificities ranging from 89% to >97% 120 , 123 and, therefore, are not recommended. However, mathematical modelling has shown that the sensitivity required for POCTs to be effective may be lower in settings where there is a high risk for transmission because treatment is delayed pending testing results or patients do not return for treatment 124 . The Xpert CT/NG assay has been successfully implemented as a near-POCT in areas such as Papua New Guinea, South Africa and remote regions of Australia 6 , 101 , 115 , 125 , 126 . However, this test is expensive, needs substantial electricity, and results take ~90 min.
Screening and prevention
Screening general populations for gonococcal infections is not indicated. However, screening or opportunistic testing can be considered for individuals at higher risk of gonococcal infection. These populations include the sexually active youth, sexual contacts of individuals having a suspected gonococcal infection, MSM, individuals with new or multiple sexual partners, individuals with HIV infection or a history of STIs, sex workers and their sexual partners, and women (≤35 years of age) and men (≤30 years of age) at initial admission to a correctional facility 6 , 13 , 83 , 127 , 128 . The US Centers for Disease Control and Prevention guidelines recommend annual screening for gonorrhoea of all sexually active females <25 years of age and older women at increased risk of infection, and screening should also be offered to young MSM 127 , 128 . More recently, in the USA, owing to observed high rates of incident infections, screening for gonorrhoea and other bacterial STIs ( C. trachomatis infections and syphilis) has been recommended at 3–6-month intervals for individuals receiving HIV PrEP 129 . In other high-income settings, there are no screening recommendations for the general population owing to the low cost-effectiveness and low population prevalence of gonorrhoea, which results in low positive predictive values of the testing and increased probability of false positive results, which could cause considerable harm for patients and their partners. No aetiologically based screening is performed in any low-income settings.
Main prevention efforts include education regarding symptomatic and asymptomatic gonorrhoea and other STIs; promotion of safe sexual behaviours (for example, increased condom use through condom-promotion education and campaigns); behaviour change communication programmes (for example, promoting fewer unknown, casual and unprotected sexual contacts and early health-seeking behaviour); improved sexual partner notification and treatment; and expansion of targeted interventions, including screening in some settings for vulnerable populations (sex workers, MSM, adolescents and patients with STIs and their sexual partners) 130 .
Given the threat of untreatable gonorrhoea due to the spread of AMR and the high burden of gonorrhoea worldwide, the need for a gonococcal vaccine has become increasingly urgent 131 , 132 , 133 . Prior to the 1990s, four vaccine candidates progressed to clinical trials: a whole cell vaccine, a partially autolysed vaccine, a pilus-based vaccine and a PorB-based vaccine 133 , 134 , 135 ; none provided much protection from infection. Gonococcal vaccine development is complicated by the biology of the gonococcus. Limitations include the scarce adaptive immune responses to gonococcal infections, lack of known correlates of protection, antigenic variability of the potential vaccine candidate antigens, production of blocking antibodies (which upon binding their target prevent the binding of other antibodies — for example, bactericidal antibodies — to the same target or other targets in close proximity) to conserved antigens, and lack of robust, small laboratory animals for testing vaccines 132 , 134 .
However, recently, it has been noted in several countries that there was a decline in the number of gonorrhoea cases following the use of meningococcal group B OMV vaccines against N. meningitidis 136 . One of these vaccines, with the trade name MeNZB, was associated with reduced rates of gonorrhoea diagnosis and of hospitalization from gonorrhoea 136 , and it seems to provide proof-of-principle to inform the development of gonococcal vaccines 137 , 138 . Research to elucidate the specific or nonspecific antigens and mechanisms involved in the MeNZB-mediated protection against gonorrhoea is crucial. MeNZB is no longer available; however, the licensed, four-component meningococcal group B vaccine 4CMenB (trade name BEXSERO; GlaxoSmithKline) includes the same OMV as MeNZB and three recombinant meningococcal antigens (Neisserial heparin-binding antigen, factor H-binding protein and Neisseria adhesin A), which are also relatively conserved compared with their gonococcal homologues 139 . Accordingly, high coverage of the 4CMenB in the population may also decrease gonorrhoea prevalence. Recently, research has exploited OMVs from N. meningitidis expressing factor H-binding protein and found that serum bactericidal antibodies against the gonococcus were produced in mice, although sera from humans immunized with 4CMenB were not bactericidal for N. gonorrhoeae 140 . These findings, together with the immunobiology research (including on N. gonorrhoeae immune suppressive responses and how they can be overcome), antigen discovery and animal modelling, are promising for vaccine development.
Management principles
Gonorrhoea is a community-based infection and often there is limited follow-up after treatment. Prompt and effective treatment reduces complications and eliminates transmission of the infection 128 . Since there are no vaccines, and host immunity cannot prevent reinfection, eradication of infections is solely reliant upon case finding and ideally microbiological diagnosis coupled with effective antimicrobial treatment 128 . Of note, because gonorrhoea also amplifies the risk for acquisition and transmission of HIV, gonorrhoea control also contributes to global efforts to reduce HIV infections. The goal of gonorrhoea management is to quickly and accurately identify infected individuals, enabling provision of timely treatment to prevent complications and transmission of infection to sexual partners and, for pregnant women, to children at the time of birth. Factors influencing management include considerations of the clinical manifestations, the disproportionate morbidity for women (PID, infertility, ectopic pregnancy and chronic pelvic pain), and stigma associated with STIs. As the infection is most common in resource-limited settings (even in high-income nations gonorrhoea is most common among marginalized populations who may have limited resources and/or limited access to health care), costs of both diagnosis and treatment may also influence the translation of management principles into practice.
Because gonorrhoea transmission most often is a consequence of sex with a person who is unaware of their infection, notification, testing and treatment of recent sexual partners is a crucial part of gonorrhoea management within communities 82 , 141 . Notification and referral of exposed sexual partners of individuals with STIs (by health-care providers, public health specialists or the partner themself) has been recommended since at least the 1940s 142 . However, programmes promoting notification of sexual partners have often proved resource intensive and failed to successfully lead to treatment of many sexual partners, probably in part owing to stigma and embarrassment regarding having an STI. Thus, ‘expedited partner therapy’ or ‘partner-delivered therapy’ (that is, the partner(s) of a patient with gonorrhoea receives oral, single dose antimicrobials delivered by the patient, without have being examined or tested) for gonococcal and chlamydial infections has been increasingly practiced in the USA with good results 143 . Currently, cefixime plus azithromycin is used for expedited partner therapy for heterosexual men and women 128 . However, this approach has raised concerns about the lack of clinical examination, lack of testing for additional STIs, lack of opportunities to trace ‘downstream’ sex partners, possible antimicrobial allergy or adverse events experienced by the partner(s) and AMR emergence.
Antimicrobial therapy
Syndromic management of urethral discharge in men can be relatively effective for gonorrhoea 116 . However, appropriate, local and aetiologically based studies to regularly refine the syndromic management algorithm(s) are imperative, and nevertheless some infections (for example, C. trachomatis and Mycoplasma genitalium infections) cannot be distinguished from gonorrhoea, resulting in overtreatment. Syndromic management of vaginal discharge both fails to detect and treat the substantial proportion of asymptomatic infections in women (who might continue to transmit the infection) and leads to vast overtreatment of symptomatic women who do not have gonorrhoea but who do have C. trachomatis , M. genitalium or Trichomonas vaginalis infection or bacterial vaginosis 109 , 116 .
Single-dose, directly observed systemic therapy (as topical therapy has not proved effective) that is provided in the care setting is preferred to ensure medications are delivered. Dual antimicrobial therapy (mainly parenteral ceftriaxone plus oral azithromycin) is currently recommended for empirical first-line therapy by the WHO global guidelines 109 and in most high-income countries, including European countries 82 , the USA 128 , Canada 144 and Australia 145 ; however, in some countries (for example, Japan 146 and, since 2019, the UK 114 ) ceftriaxone high-dose (1 g) monotherapy is recommended 147 , 148 , 149 . In some international and national guidelines, cefixime plus azithromycin is recommended as an alternative regimen, but only if ceftriaxone is not available or the injection is refused 82 , 128 . There is an ongoing debate among experts as to whether single or dual antimicrobial therapy should be the recommended therapy for uncomplicated gonorrhoea. The rationale for introducing dual therapy was to address the problem of C. trachomatis co-infection, which occurs in 10–40% of individuals with urogenital gonorrhoea 150 , as well as a hypothetical benefit of reducing the emergence and/or spread of AMR (particularly resistance to ceftriaxone) in N. gonorrhoeae . When possible, well tolerated oral therapy is preferred by both patients and clinicians 151 . Finally, individuals with gonorrhoea are often co-infected with other pathogens, including C. trachomatis , T. vaginalis , Treponema pallidum and/or M. genitalium and, therefore, require treatment either with agents that are also effective against these pathogens or with co-therapy.
The continuing development of AMR by the gonococcus, coupled with a diminished pipeline for the development of new antimicrobials have narrowed available therapies for gonorrhoea to a single agent that is sufficiently effective for first-line monotherapy (that is, parenteral ceftriaxone 16 , 152 ), which is frequently given together with azithromycin. If ceftriaxone is unavailable, the patient has β-lactam antimicrobial allergy or the patient is infected with a ceftriaxone-resistant gonococcal strain, therapy is challenging and highly variable, often requiring ciprofloxacin monotherapy (if the gonococcal strain causing the infection has been proven susceptible by phenotypic or genetic resistance testing 82 , 114 ), high-dose (2 g) azithromycin monotherapy, spectinomycin (together with high-dose azithromycin, particularly if pharyngeal gonorrhoea has not been excluded) or gentamicin (together with high-dose azithromycin, particularly if pharyngeal gonorrhoea has not been excluded) 82 , 128 . However, each of these alternate therapies has limitations related to gonococcal resistance, antimicrobial availability and/or patient tolerance. Progressive decreases in susceptibility of N. gonorrhoeae to ceftriaxone, as well as to other antimicrobials, create a pressing need for continued monitoring of gonococcal AMR through surveillance networks such as the WHO Global Gonococcal Antimicrobial Surveillance Programme (WHO GASP) 15 , 153 , the European GASP (Euro-GASP) 154 , 155 , 156 and the US Centers for Disease Control and Prevention Gonococcal Isolate Surveillance Project (GISP) 157 , 158 ; Euro-GASP and GISP additionally collect clinical and epidemiological data on the corresponding patients.
Practical applications
Gonorrhoea remains a global public health threat. The biological characteristics of N. gonorrhoeae and its proven propensity to develop AMR, the varied clinical manifestations of the infection that may not be obvious or pathogen-specific (particularly for women and extragenital infections), and the limited resources that are dedicated to gonorrhoea control all contribute to the limited success of present gonorrhoea control efforts. Therapy may be hindered by the lack of recommended, high-quality antimicrobials. Current main reliance on only one consistently effective antimicrobial (injectable ceftriaxone) may make effective treatment difficult. Perceptions by patients that they may be resistant or allergic to β-lactam antimicrobials, including ceftriaxone, the logistical constraints of parenteral therapy and fear/avoidance of injections may result in the use of less-effective oral therapy. Therapy is also limited in some regions by suboptimal or complete absence of surveillance of infection and particularly AMR, leading to treatment with antimicrobials that are ineffective. Although improved surveillance has increased appreciation of the threat of AMR, this surveillance is not fully representative, being insufficient or even lacking in areas where the infection is most common 15 , 153 , 159 .
On the policy level, limited health-care resources directed towards this public health problem (in low-income and middle-income nations and even in high-income nations) have created a tension between diagnostic test cost and ensuring a ready supply of medications for gonorrhoea control. The cost of paying for diagnostic testing may erode the funds available for therapy, thereby forcing public health officials to prioritize screening initiatives. In recent years, clinical microscopy (Gram stain) as a low-cost POCT has become less available, owing to the lack of availability of microscopes and adequate technical training in the methodology.
All these challenges are sometimes amplified by social factors. Stigma is a pervasive and powerful force that affects the prioritization of gonorrhoea as a public health problem and influences the behaviour of individuals with, or at risk for, gonorrhoea with regard to health care-seeking behaviour and partner notification. Stigma also affects health-care provider attitudes and practices, including evaluation of STI risk and appropriate screening 159 .
At the individual level, few individuals wish to identify themselves as being at risk for STIs, potentially inhibiting discussion of STI risk with their health-care provider, prevention measures and seeking evaluation for genitourinary symptoms and signs. Limited access to health care may also prevent or delay recommended STI screening or evaluation of symptoms when present. Finally, individuals diagnosed with gonorrhoea or other STIs may fail to notify their sex partners of their risk of infection, thereby increasing the probability of complications or continuing transmission.
Clinicians too are sometimes hindered by perceived social factors in evaluating and managing individuals with or at risk for STIs. Busy clinicians may assume that their patients are not at risk or hesitate to take sexual histories without a cue to action from their patients, such as a history of possible exposure or genitourinary symptoms or signs, worrying that to ask such questions might be offensive to patients, when data in fact indicate that, if properly presented, this is not the case 159 . Clinician reticence, along with individual embarrassment and/or shame may also hinder partner notification.
Thus, although the principles of gonorrhoea management are well known, there are numerous areas within the current management strategies that need to be improved.
Quality of life
As gonorrhoea is an STI, its diagnosis is often associated with perceptions of social stigma, shame and denial, and can lead to intense embarrassment and fear of retaliation, domestic violence or loss of relationships, including marriages 160 . In the 1960s, the sociologist Erving Goffman described stigma as “undesired differentness” and “discrediting” 161 — a finding reinforced by research findings in the 1990s showing that STI-related stigma resulted in lower testing rates for gonorrhoea 162 . More recent studies have shown that stigma in different populations contributes to a reduction in seeking testing for STIs, reluctance to notify sexual partners and lower levels of treatment compliance 163 , 164 . For example, in Bhutan, perceived stigma was identified as a key reason for high levels (>50%) of loss to follow-up among patients diagnosed with gonorrhoea 165 . Research found that common coping strategies among people with gonorrhoea in an urban American setting included denial and disengagement — although these behaviours did not greatly affect rates of partner notification 166 . These findings, specific to gonorrhoea, are illustrative of more general findings that stigma influences STI care-seeking. Research noted a reluctance to seek STI testing in young women from socioeconomically marginalized neighbourhoods in Canada, owing to “stigma and the fear of being ostracized” 167 and studies found that, among African–American men, increasing STI-related stigma was “significantly associated with…decreased odds of having been tested, [and]…decreased willingness to notify non-main partners” 168 ; these factors may contribute to the observed disparities in the distribution of STIs across the intersectional inequalities of ethnicity and gender 169 . In Tigray, Ethiopia, rates of loss to follow-up were lower among patients with low levels of STI-related stigma than in study participants reporting high levels of stigma 164 .
At the policy level, stigma around gonorrhoea probably contributes to the widespread lack of attention and resource allocation within public health global and national programmes. A recent review of the challenges and opportunities for STI control argued that stigma associated with gonorrhoea and other STIs arises, in part, from ‘condemnatory moral attitudes’ around the behaviours leading to risk of infection — in particular same-sex relationships and transactional sex 170 . Earlier research investigating gonorrhoea control in the USA in the 1970s and 1980s similarly argued that “society’s propensity to view gonorrhoea as a disease of ‘immoral’ people” directly contributed to the lack of resources and attention paid to the infection 171 . Qualitative research on the lack of political prioritization afforded to STI control in China confirmed that STIs received a lower place on the health agenda than HIV infection, as decision makers associated them with ‘immorality’ and patients were considered ‘condemnable’ 172 .
Arguably, the high levels of stigma and accompanying negative framing of gonorrhoea and other STIs exert the most substantial effect on quality of life measures associated with gonorrhoea. Perceptions of embarrassment and humiliation that a diagnosis may bring — both for the affected individuals and their sexual partners — combined with under-resourced public health control programmes, contribute to undiagnosed or poorly treated infections, thereby increasing risks of onward transmission and individual clinical complications and longer-term sequelae caused by this otherwise treatable infection.
Paradoxically, the rise of AMR in N. gonorrhoeae may, potentially, force policy-level decision makers to act to devote more attention to the prevention and control of gonorrhoea. However, it should be emphasised that interventions to tackle gonococcal AMR are only likely to succeed if they address not only questions of appropriate antimicrobial use/misuse, but also aim to decrease the global burden of gonorrhoea, which also requires reducing the perception of associated shame and stigma. Effective interventions to decrease stigma and increase patient quality of life should be directed not only at individual and community levels, but also at the political level, to identify and address the social conditions giving rise to stigma and promote institutional fairness 173 .
It is imperative to address many global issues for the successful management and control of gonorrhoea. These key priorities and research efforts span all fields, from epidemiology of the pathogen and the disease to the quality of life of patients (Box 2 ). Of note, reducing the perception of shame, humiliation and stigma that is associated with a diagnosis of gonorrhoea and with certain sexual orientations (for example, MSM) in many settings is crucial to obtain more accurate incidence and prevalence data and to decrease the global burden of gonorrhoea, which would also substantially reduce gonococcal AMR. Effective interventions to decrease STI-associated stigma should be implemented at individual and community levels, and at the social and political levels where social conditions giving rise to stigma should be identified and tackled 173 . Gonorrhoea and other STIs need to be considered and managed by individuals, the health system, the general community and at the political level in all countries in recognition of the right to health services free of discrimination and without stigma.
Box 2 Key priorities in gonorrhoea research and control
Decreasing the perception of stigma, humiliation and shame associated with gonorrhoea and other sexually transmitted infections (STIs), and ensuring that services and interventions are delivered free of discrimination, leaving no populations behind
Increasing knowledge of the incidence and prevalence of the infection and its complications and sequelae in the general population and subpopulations
Expanding global antimicrobial resistance (AMR) surveillance (phenotypic and genetic AMR testing), including surveillance of treatment failures and antimicrobial use/misuse, in combination with whole-genome sequencing and clinical and epidemiological data from patients
Improving knowledge of the natural course and pathogenesis, including genomic, physiological, pathogenic and virulence mechanisms of Neisseria gonorrhoeae in different anatomical sites and understanding the emergence, evolution, spread and biological costs or benefits (fitness) of AMR
Understanding of pharmacokinetics and pharmacodynamics of current and future therapeutic antimicrobials in urogenital and particularly extragenital sites, to inform treatment guidelines
Increasing diagnostic testing (also to detect asymptomatic gonorrhoea), increasing use of validated and quality-assured nucleic acid amplification tests and developing rapid, appropriate and affordable point-of-care tests, which should also enable simultaneous prediction of AMR or susceptibility status
Strengthening prevention (for example, increasing the use of condoms and of out-of-box approaches, such as the use of antiseptic mouthwash to prevent acquisition and transmission of pharyngeal gonorrhoea 215 )
Improving the understanding of the effects of pre-exposure prophylaxis on the prevalence of gonorrhoea and other STIs in different populations, the risk factors involved, and the ideal counselling, monitoring and screening intervals for individuals taking pre-exposure prophylaxis
Developing gonococcal vaccine(s), for which substantial progress has been made in recent years 131 , 132 , 133 , 134 , 136 , 137 , 138 , 189 , 191 , 216
Promoting early diagnosis and treatment of patients and their partners, following evidence-based international and national guidelines
Promoting responsible antimicrobial use and stewardship (both STI-related and on a population level), as excessive antimicrobial use can decrease the susceptibility of N. gonorrhoeae to therapeutic drugs, both directly (through selection of AMR in N. gonorrhoeae ) and indirectly (through selection of AMR determinants in, for example, commensal Neisseria spp. that are subsequently shared through horizontal genetic transfer with N. gonorrhoeae 217 )
Developing novel therapeutic antimicrobials and strategies to preserve the efficacy of current and future antimicrobials
The incidence of gonorrhoea is increasing, especially in high-income settings globally. However, global population-based incidence and prevalence data are extremely scarce from most settings and, even in high-income settings where surveillance is conducted in a more systematic and regular manner, the surveillance data should be interpreted with caution as the surveillance systems, diagnostic testing, methodologies and quality assurance are not standardized across countries and remain weak in several settings 33 , 36 . Additionally, the current prevalence of serious complications and sequelae due to gonorrhoea is mainly unknown and estimates are mostly based on historical data. WGS will revolutionize our understanding of the molecular epidemiology (that is, the geographical and temporal spread) of N. gonorrhoeae strains. WGS is substantially more accurate than previously used molecular epidemiological typing methods and can adequately describe the emergence, transmission and evolution of AMR gonococcal strains both geographically and temporally, as well as predict AMR with adequate accuracy 45 , 156 , 174 , 175 , 176 , 177 , 2 mg/L) Neisseria gonorrhoeae isolates in Europe from 2009 to 2014. J. Antimicrob. Chemother. 71, 3109–3116 (2016)." href="#ref-CR178" id="ref-link-section-d126509203e5547_4">178 , 179 , 180 , 181 , 182 , 183 , 184 . However, it is important to strongly emphasize that the full benefits of using WGS for both molecular and infection epidemiology can only be achieved if the WGS data are linked to phenotypical data for the gonococcal isolates and the clinical and epidemiological data for the corresponding patients with gonorrhoea. Notably, WGS of gonococcal isolates with joint analysis of clinical and epidemiological data has also already been introduced and provided increased understanding of, for example, the distribution of AMR and susceptible gonococcal strains in different populations nationally and regionally in the international Euro-GASP (which currently includes 27 European countries) 156 .
Our understanding of the pathophysiology of gonorrhoea is still limited in many areas, especially the natural course of the infection (including duration and spontaneous resolution), the dynamics of pathogenesis and infection (such as transmission, average time to detection and treatment in different populations, effects of treatment (or co-treatment for other concomitant STIs) on innate and adaptive immunity, host damage and possible host protection) and immune responses and their suppression in urogenital and particularly extragenital sites, such as the pharynx. Improving the knowledge in these areas would enable us to more effectively use mathematical modelling in the gonorrhoea and gonococcal AMR field, taking into account microbiological, genomic, evolutionary, clinical, immunological and epidemiological data 185 , as well as in vaccine development.
After the introduction of any new therapeutic antimicrobial for gonorrhoea, N. gonorrhoeae has rapidly acquired or developed decreased susceptibility or resistance to it (Fig. 1 ) via several AMR mechanisms: enzymatic destruction or modification of the antimicrobial, modification or protection of antimicrobial targets to avoid binding, increased export of the antimicrobial (for example, through the MtrC–MtrD–MtrE efflux pump) and decreased uptake of the antimicrobial (for instance, through the porin PorB) 16 . Some AMR determinants, particularly target alterations, directly cause AMR, whereas others cannot result in AMR on their own and require the presence of additional AMR determinants. The accumulation of many AMR determinants does not seem to substantially reduce the biological fitness of N. gonorrhoeae 16 , 17 , 18 , 19 , 20 , 21 , and some AMR determinants seem to even enhance the fitness of specific gonococcal strains 19 , 20 , 21 . Nevertheless, we need to substantially improve our understanding and definition of fitness as well as of compensatory mutations that could restore possible fitness cost in N. gonorrhoeae . We need detailed knowledge regarding how gonococcal AMR determinants affect the fitness of gonococcal strains, how fitness affects the emergence and spread of AMR strains and how these strains become established in the circulating gonococcal populations. Thus, we need to investigate how the fitness of AMR strains may affect the competition with wild-type antimicrobial susceptible strains (which is mainly the current fitness definition in microbiological research) and its effects on several other factors, such as transmissibility, duration of infection in different anatomical sites and proportion of symptomatic and asymptomatic infections and severe complications and sequelae in heterogeneous populations with different sexual behaviours. Further research is also needed to identify and characterize in detail known or novel AMR determinants in clinical gonococcal isolates (including their induction and selection, evolution, effect on AMR and biological fitness) and to develop and evaluate genetic AMR prediction tests that can supplement the culture-based AMR surveillance.
In many settings, mostly in less-resourced areas (in which frequently the prevalence of gonorrhoea is the highest), the diagnosis, testing, case reporting and prevention of gonorrhoea remain suboptimal. Thus, it is important to widely implement the use of cost-effective, appropriate and quality-assured NAATs. If required, these NAATs can be performed in centralized reference laboratories for cost-effectiveness and to maintain a high level of quality assurance. In addition, rapid, appropriate POCTs for the diagnosis of gonorrhoea and other STIs are urgently needed. Gonococcal POCTs should ideally simultaneously predict AMR to inform treatment. For some antimicrobials, such as ciprofloxacin, mathematical modelling has indicated that POCTs with high sensitivity to detect AMR can be more effective than NAATs and even culture to preserve the effectiveness of the antimicrobial. By contrast, POCTs detecting N. gonorrhoeae without reliable AMR detection may accelerate the spread of AMR gonococcal strains 186 . Several rapid, sensitive and specific NAAT-based POCTs for gonorrhoea are in the pipeline and will be available in the coming few years 101 , 122 , 170 , 187 (Table 3 ). Accordingly, it will soon be essential to prepare health-care systems for use of these POCTs by including them in STI training modules, management guidelines, diagnostic algorithms and regulatory frameworks. Limitations to the adoption of POCTs are considerable and include time for results; cost of the instrument; lack of required infrastructure, quality assurance and reporting criteria; supply chain issues that may discourage use; lack of clear recommendations on the inclusion of POCTs in diagnostic algorithms and regulatory frameworks, lack of training opportunities and education of health-care workers about the utility and advantages of POCTs; and worries by laboratory-based personnel that out-of-laboratory testing may infringe on job security 118 .
In an era of high prevalence of AMR in N. gonorrhoeae coupled with the widespread use of diagnostic gonococcal NAATs internationally, it is essential to retain and strengthen the ability to perform gonococcal culture, which is the only method that enables complete AMR testing, because surveillance of gonococcal AMR (preferably minimum inhibitory concentration (MIC)-based) and ideally also of cases of treatment failure is imperative. In settings where NAATs solely are used for the diagnosis of gonorrhoea, participation in organized and quality-assured national, regional and/or international GASPs is crucial.
WGS and other new technologies such as transcriptomics and proteomics are also informing the development of N. gonorrhoeae diagnostics and vaccines 156 , 174 , 175 , 176 , 177 , 2 mg/L) Neisseria gonorrhoeae isolates in Europe from 2009 to 2014. J. Antimicrob. Chemother. 71, 3109–3116 (2016)." href="#ref-CR178" id="ref-link-section-d126509203e5654_4">178 , 179 , 180 , 181 , 182 , 183 , 188 , 189 , 190 , 191 . For developing gonococcal vaccines, a number of promising protein antigens have been described and characterized, including proteins involved in colonization (for example, PilC, PilQ, PorB, Opa and OmpA), evasion of innate defences (for example, MtrE, SliC, Ng-ACP, MsrAB, Lst and PorB) and nutrient acquisition (for example, TbpA, TbpB, LbpA and LbpB); structural proteins (for example, BamA, BamE, NGO2054 and NGO2111); other proteins such as AniA (implicated in nitrate reduction) and MetQ (methionine transporter that promotes survival in macrophages); the 2C7 epitope (peptide mimetic of LOS epitope); and OMVs 131 , 132 , 134 . Many of the promising new vaccine targets for N. gonorrhoeae have been identified through proteomic approaches and transcriptome analysis of genes expressed during gonococcal infections 188 , 189 , 190 , 192 . Furthermore, to overcome the restrictions of the current model of female mice treated with 17β-oestradiol, new animal models for N. gonorrhoeae infection are being developed, such as transgenic mice that mimic human infections and express human cell adhesion molecules or iron-binding molecules 193 , 194 , and a transgenic mice model expressing human complement factor H is available for the closely related N. meningitidis 195 .
Currently available genetic assays have shortcomings (such as cross-reactions with nongonococcal Neisseria species in clinical, particularly pharyngeal, specimens, and suboptimal sensitivity and/or specificity) that limit their prediction of resistance or susceptibility to currently recommended therapeutic antimicrobials (except for ciprofloxacin, for which the sensitivity and specificity of NAATs are generally >95%); additionally, newly emerging AMR determinants are not detected 196 , 197 , 198 . However, future improved rapid POCTs that detect both N. gonorrhoeae and its resistance or susceptibility to several antimicrobials will guide individualized therapy at the first health-care visit and restrict the use of last-line antimicrobials 196 , 197 , 198 , 199 . Such POCTs will improve the management and control of both gonorrhoea and N. gonorrhoeae AMR. WGS can also be used for prediction of AMR and MICs of antimicrobials with reasonably high accuracy 156 , 182 , 183 , 184 . Rapid, real-time sequencing with the hand-held MinION sequencer was shown to generate fairly accurate genome sequences and be able to predict resistance to ciprofloxacin and azithromycin and decreased susceptibility or resistance to cefixime in N. gonorrhoeae 183 . The rapid development of WGS technologies with decreasing complexity and cost and faster turnaround times may make these technologies suitable for N. gonorrhoeae detection and prediction of resistance or susceptibility to therapeutic antimicrobials at the diagnostic setting, including at point-of-care.
The global issue of AMR in N. gonorrhoeae will probably continue to escalate, and we cannot rely on the last-line ceftriaxone (plus azithromycin) indefinitely. Consequently, new antimicrobials, with novel mechanisms of action, for monotherapy and/or inclusion in dual therapies for urogenital and extragenital gonorrhoea are crucially needed. Some recently developed new antimicrobials, namely, the spiropyrimidinetrione zoliflodacin 200 , 201 , 202 , 203 , 204 and triazaacenaphthylene gepotidacin 205 , 206 , 207 , will both soon be in phase III randomized clinical controlled trials for uncomplicated gonorrhoea. Additional promising novel antimicrobials in earlier development that deserve further attention for the treatment of gonorrhoea (and possibly additional STIs) are, for example, lefamulin 208 , 209 and SMT-571 (ref. 210 ). However, until novel antimicrobials are available, it is imperative to increase our knowledge regarding ideal treatment, including dosing regimens, of gonorrhoea and other STIs, such as C. trachomatis and M. genitalium infections, with the available antimicrobials ceftriaxone, azithromycin and doxycycline. Clearly, a more holistic view on the treatment of bacterial STIs and understanding the effect of any new bacterial STI treatment on other STI pathogens and the bystander microbiota is essential. Current knowledge regarding the pharmacokinetics and pharmacodynamics of the available antimicrobials in the treatment of gonorrhoea and other STIs at urogenital and particularly extragenital sites is highly limited 211 and requires substantially increased attention to inform ideal dosing regimens, and multiple dose regimens for gonorrhoea might be required.
Edwards, J. L., Shao, J. Q., Ault, K. A. & Apicella, M. A. Neisseria gonorrhoeae elicits membrane ruffling and cytoskeletal rearrangements upon infection of primary human endocervical and ectocervical cells. Infect. Immun. 68 , 5354–5363 (2000).
Article CAS PubMed PubMed Central Google Scholar
Evans, B. A. Ultrastructural study of cervical gonorrhea. J. Infect. Dis. 136 , 248–255 (1977).
Article CAS PubMed Google Scholar
Barlow, D. & Phillips, I. Gonorrhoea in women. Diagnostic, clinical, and laboratory aspects. Lancet 1 , 761–764 (1978).
Schmale, J. D., Martin, J. E. Jr & Domescik, G. Observations on the culture diagnosis of gonorrhea in women. JAMA 210 , 312–314 (1969).
Quillin, S. J. & Seifert, H. S. Neisseria gonorrhoeae host adaptation and pathogenesis. Nat. Rev. Microbiol. 16 , 226–240 (2018). This review discusses sex-related symptomatic gonorrhoea and provides a detailed overview of the bacterial factors, on molecular levels, that are important for the different stages of pathogenesis, including transmission, colonization and immune evasion .
Elias, J. F. & Vogel, U. in Manual of Clinical Microbiology 12th edn Vol. 1(eds Carroll, C. C. et al.) 640–655 (American Society for Microbiology, 2019).
Adeolu, M. & Gupta, R. S. Phylogenomics and molecular signatures for the order Neisseriales: proposal for division of the order Neisseriales into the emended family Neisseriaceae and Chromobacteriaceae fam. nov. Antonie van Leeuwenhoek 104 , 1–24 (2013).
Article PubMed Google Scholar
Tønjum, T. & van Putten, J. in Infectious Diseases 4th edn (eds Cohen, J., Powderly, W. G. & Steven M. Opal) 1553-1564 (Elsevier, 2016).
Liu, G., Tang, C. M. & Exley, R. M. Non-pathogenic Neisseria: members of an abundant, multi-habitat, diverse genus. Microbiology 161 , 1297–1312 (2015).
Johnson, A. P. The pathogenic potential of commensal species of Neisseria. J. Clin. Pathol. 36 , 213–223 (1983).
Seifert, H. S. Location, location, location — commensalism, damage and evolution of the pathogenic Neisseria. J. Mol. Biol 431 , 3010–3014 (2019).
Hook, E. W. 3rd & Handsfield, H. H. in Sexually Transmitted Diseases (eds Holmes, K. K. et al.) 4th edn, 627–645 (McGraw-Hill Education, 2008). This comprehensive chapter describes different clinical manifestations of gonorrhoea .
Public Health Agency of Canada. Canadian Guidelines on Sexually Transmitted Infections — Management and treatment of specific infections — Gonococcal Infections (Government of Canada, Ottawa, 2013) (modified Sept 2017).
World Health Organization. Global Action Plan to Control the Spread and Impact of Antimicrobial Resistance in Neisseria gonorrhoeae (World Health Organization, 2012).
Wi, T. et al. Antimicrobial resistance in Neisseria gonorrhoeae : global surveillance and a call for international collaborative action. PLoS Med. 14 , e1002344 (2017).
Article PubMed PubMed Central CAS Google Scholar
Unemo, M. & Shafer, W. M. Antimicrobial resistance in Neisseria gonorrhoeae in the 21st century: past, evolution, and future. Clin. Microbiol. Rev. 27 , 587–613 (2014). This review provides an extensive overview regarding gonorrhoea treatment regimens and emerging antimicrobial resistance, including genetic and phenotypic AMR determinants .
Wadsworth, C. B., Arnold, B. J., Sater, M. R. A. & Grad, Y. H. Azithromycin resistance through interspecific acquisition of an epistasis-dependent efflux pump component and transcriptional regulator in Neisseria gonorrhoeae . mBio 9 , e01419–18 (2018).
Rouquette-Loughlin, C. E. et al. Mechanistic basis for decreased antimicrobial susceptibility in a clinical isolate of Neisseria gonorrhoeae possessing a mosaic-like mtr efflux pump locus. mBio 9 , e02281–18 (2018).
Article PubMed PubMed Central Google Scholar
Kunz, A. N. et al. Impact of fluoroquinolone resistance mutations on gonococcal fitness and in vivo selection for compensatory mutations. J. Infect. Dis. 205 , 1821–1829 (2012).
Warner, D. M., Folster, J. P., Shafer, W. M. & Jerse, A. E. Regulation of the MtrC-MtrD-MtrE efflux-pump system modulates the in vivo fitness of Neisseria gonorrhoeae . J. Infect. Dis. 196 , 1804–1812 (2007).
Warner, D. M., Shafer, W. M. & Jerse, A. E. 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. 70 , 462–478 (2008).
Rowley, J. et al. Chlamydia, gonorrhoea, trichomoniasis and syphilis: global prevalence and incidence estimates, 2016. Bull. World Health Organ. 97 , 548–562P (2019).
Adler, M., Foster, S., Richens, J. & Slavin, H. Sexual Health and Care. Sexually Transmitted Infections, Guidelines for Prevention and Treatment . Health and Population Division Occasional Paper 136 (Overseas Development Administration, London, 1996).
Aral, S. O. et al . in Sexually Transmitted Diseases , 4th edn (eds Holmes, K. K. et al.) 54–92 (McGraw-Hill, 2008).
Dallabetta, G. A., Laga, M. & Lamptey, P. R. Control of Sexually Transmitted Diseases: A Handbook for the Design and Management of Programs (AIDSCAP/Family Health International, 1996).
Aral, S. O., Fenton, K. A. & Holmes, K. K. Sexually transmitted diseases in the USA: temporal trends. Sex. Transm. Infect. 83 , 257–266 (2007).
Fenton, K. A. & Lowndes, C. M. Recent trends in the epidemiology of sexually transmitted infections in the European union. Sex. Transm. Infect. 80 , 255–263 (2004).
Mohammed, H. et al. 100 years of STIs in the UK: a review of national surveillance data. Sex. Transm. Infect. 94 , 553–558 (2018).
Centers for Disease Control and Prevention. Tracking the hidden epidemics, trends in STDs in the United States 2000. CDC www.cdc.gov/std/trends2000/trends2000.pdf (2000).
Centers for Disease Control and Prevention. STDs in men who have sex with men. CDC https://www.cdc.gov/std/stats17/msm.htm . (2017).
Centers for Disease Control and Prevention. New CDC analysis shows steep and sustained increases in STDs in recent years. CDC https://www.cdc.gov/media/releases/2018/p0828-increases-in-stds.html (2018).
Centers for Disease Control and Prevention. Gonorrhea. CDC https://www.cdc.gov/std/stats17/gonorrhea.htm (2017).
European Centre for Disease Prevention and Control Surveillance Atlas of Infectious Diseases. Surveillance atlas of infectious diseases. ECDC https://www.ecdc.europa.eu/en/surveillance-atlas-infectious-diseases (2017).
Public Health England. Health Protection Report volume 12 issue 20: news (8 June). PHE https://www.gov.uk/government/publications/health-protection-report-volume-12-2018/hpr-volume-12-issue-20-news-8-june (2018).
Torrone, E. A. et al. Prevalence of sexually transmitted infections and bacterial vaginosis among women in sub-Saharan Africa: an individual participant data meta-analysis of 18 HIV prevention studies. PLoS Med. 15 , e1002511 (2018).
Dehne, K. L. et al. A survey of STI policies and programmes in Europe: preliminary results. Sex. Transm. Infect. 78 , 380–384 (2002).
Kojima, N., Davey, D. J. & Klausner, J. D. Pre-exposure prophylaxis for HIV infection and new sexually transmitted infections among men who have sex with men. AIDS 30 , 2251–2252 (2016).
Traeger, M. W. et al. Association of HIV preexposure prophylaxis with incidence of sexually transmitted infections among individuals at high risk of HIV infection. JAMA 321 , 1380–1390 (2019).
World Health Organization. Prevention and control of seually transmittted infections (STIs) in the era of oral pre-exposure prophylaxis (PrEP) for HIV. Technical Brief. WHO https://apps.who.int/iris/bitstream/handle/10665/325908/WHO-CDS-HIV-19.9-eng.pdf?ua=1 (2019).
Celum C. Oral pre-exposure prophylaxis (PrEP) for prevention [MOSA3401]. 22nd International AIDS Conference (AIDS 2018) http://programme.aids2018.org/People/PeopleDetailStandalone/7599 (2018).
McCormack, S. et al. Pre-exposure prophylaxis to prevent the acquisition of HIV-1 infection (PROUD): effectiveness results from the pilot phase of a pragmatic open-label randomised trial. Lancet 387 , 53–60 (2016).
Morse, S. A. Neisseria gonorrhoeae : physiology and metabolism. Sex. Transm. Dis. 6 , 28–37 (1979).
Rohde, K. H. & Dyer, D. W. Mechanisms of iron acquisition by the human pathogens Neisseria meningitidis and Neisseria gonorrhoeae . Front. Biosci. 8 , d1186–d1218 (2003).
Cole, J. A. Legless pathogens: how bacterial physiology provides the key to understanding pathogenicity. Microbiology 158 , 1402–1413 (2012).
Sanchez-Buso, L. et al. The impact of antimicrobials on gonococcal evolution. Nat. Microbiol. 4 , 1941–1950 (2019). This genomics paper provides evidence that the modern gonococcal population is not as old as previously anticipated and has been formed by antimicrobial treatment, leading to the emergence of one multidrug-resistant lineage and one multisusceptible lineage with different evolutionary strategies .
Tobiason, D. M. & Seifert, H. S. The obligate human pathogen, Neisseria gonorrhoeae , is polyploid. PLoS Biol. 4 , 1069–1078 (2006).
Article CAS Google Scholar
Unemo, M. et al. The novel 2016 WHO Neisseria gonorrhoeae reference strains for global quality assurance of laboratory investigations: phenotypic, genetic and reference genome characterization. J. Antimicrob. Chemother. 71 , 3096–3108 (2016).
Goodman, S. D. & Scocca, J. J. Identification and arrangement of the DNA sequence recognized in specific transformation of Neisseria gonorrhoeae . Proc. Natl Acad. Sci. USA 85 , 6982–6986 (1988).
Berry, J. L., Cehovin, A., McDowell, M. A., Lea, S. M. & Pelicic, V. Functional analysis of the interdependence between DNA uptake sequence and its cognate ComP receptor during natural transformation in Neisseria species. PLoS Genet. 9 , e1004014 (2013).
Bennett, J. S. et al. Species status of Neisseria gonorrhoeae : evolutionary and epidemiological inferences from multilocus sequence typing. BMC Biol. 5 , 35 (2007).
Maiden, M. C. et al. Multilocus sequence typing: a portable approach to the identification of clones within populations of pathogenic microorganisms. Proc. Natl Acad. Sci. USA 95 , 3140–3145 (1998).
Goire, N. et al. Mixed gonococcal infections in a high-risk population, Sydney, Australia 2015: implications for antimicrobial resistance surveillance? J. Antimicrob. Chemother. 72 , 407–409 (2017).
Martin, I. M. & Ison, C. A. Detection of mixed infection of Neisseria gonorrhoeae . Sex. Transm. Infect. 79 , 56–58 (2003).
Unemo, M. & Shafer, W. M. Antibiotic resistance in Neisseria gonorrhoeae : origin, evolution, and lessons learned for the future. Ann. NY Acad. Sci. 1230 , E19–E28 (2011).
Piekarowicz, A. et al. Characterization of the dsDNA prophage sequences in the genome of Neisseria gonorrhoeae and visualization of productive bacteriophage. BMC Microbiol. 7 , 66 (2007).
Stohl, E. A., Dale, E. M., Criss, A. K. & Seifert, H. S. Neisseria gonorrhoeae metalloprotease NGO1686 is required for full piliation, and piliation is required for resistance to H2O2- and neutrophil-mediated killing. mBio 4 , e00399–13 (2013).
Biswas, G. D., Sox, T., Blackman, E. & Sparling, P. F. Factors affecting genetic transformation of Neisseria gonorrhoeae. J. Bacteriol. 129 , 983–992 (1977).
CAS PubMed PubMed Central Google Scholar
Dehio, C., Gray-Owen, S. D. & Meyer, T. F. The role of neisserial Opa proteins in interactions with host cells. Trends Microbiol. 6 , 489–495 (1998).
Sadarangani, M., Pollard, A. J. & Gray-Owen, S. D. Opa proteins and CEACAMs: pathways of immune engagement for pathogenic Neisseria. FEMS Microbiol. Rev. 35 , 498–514 (2011).
Deo, P. et al. Outer membrane vesicles from Neisseria gonorrhoeae target PorB to mitochondria and induce apoptosis. PLoS Pathog. 14 , e1006945 (2018).
Massari, P., Ram, S., Macleod, H. & Wetzler, L. M. The role of porins in neisserial pathogenesis and immunity. Trends Microbiol. 11 , 87–93 (2003).
Madico, G. et al. Factor H binding and function in sialylated pathogenic Neisseriae is influenced by gonococcal, but not meningococcal, porin. J. Immunol. 178 , 4489–4497 (2007).
Olesky, M., Zhao, S., Rosenberg, R. L. & Nicholas, R. A. Porin-mediated antibiotic resistance in Neisseria gonorrhoeae : ion, solute, and antibiotic permeation through PIB proteins with penB mutations. J. Bacteriol. 188 , 2300–2308 (2006).
Shafer, W. M. et al. in Efflux-Mediated Antimicrobial Resistance in Bacteria (eds Li, X. Z., Elkins,C. & Zgurskaya, H.) 439-469 (Adis, Cham, 2016).
Hagman, K. E. et al. Resistance of Neisseria gonorrhoeae to antimicrobial hydrophobic agents is modulated by the mtrRCDE efflux system. Microbiology 141 , 611–622 (1995).
Lee, E. H. & Shafer, W. M. The farAB-encoded efflux pump mediates resistance of gonococci to long-chained antibacterial fatty acids. Mol. Microbiol. 33 , 839–845 (1999).
Hooper, R. R. et al. Cohort study of venereal disease. I: the risk of gonorrhea transmission from infected women to men. Am. J. Epidemiol. 108 , 136–144 (1978).
Cohen, M. S. 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 349 , 1868–1873 (1997).
Price, M. A. et al. Addition of treatment for trichomoniasis to syndromic management of urethritis in Malawi: a randomized clinical trial. Sex. Transm. Dis. 30 , 516–522 (2003).
Melly, M. A., Gregg, C. R. & McGee, Z. A. Studies of toxicity of Neisseria gonorrhoeae for human fallopian tube mucosa. J. Infect. Dis. 143 , 423–431 (1981).
Melly, M. A., McGee, Z. A. & Rosenthal, R. S. Ability of monomeric peptidoglycan fragments from Neisseria gonorrhoeae to damage human fallopian-tube mucosa. J. Infect. Dis. 149 , 378–386 (1984).
Escobar, A., Rodas, P. I. & Acuña-Castillo, C. Macrophage– Neisseria gonorrhoeae interactions: a better understanding of pathogen mechanisms of immunomodulation. Front. Immunol. 9 , 3044 (2018).
Criss, A. K. & Seifert, H. S. A bacterial siren song: intimate interactions between Neisseria and neutrophils. Nat. Rev. Microbiol. 10 , 178–190 (2012).
Massari, P., Ho, Y. & Wetzler, L. M. Neisseria meningitidis porin PorB interacts with mitochondria and protects cells from apoptosis. Proc. Natl Acad. Sci. USA 97 , 9070–9075 (2000).
Muller, A. et al. Targeting of the pro-apoptotic VDAC-like porin (PorB) of Neisseria gonorrhoeae to mitochondria of infected cells. EMBO J. 19 , 5332–5343 (2000).
Shaughnessy, J., Ram, S. & Rice, P. A. Biology of the gonococcus: disease and pathogenesis. Methods Mol. Biol. 1997 , 1–27 (2019). This review describes gonorrhoea, its epidemiology, the structure and function of major surface components involved in pathogenesis, and mechanisms that gonococci use to evade immune responses .
Densen, P. Interaction of complement with Neisseria meningitidis and Neisseria gonorrhoeae . Clin. Microbiol. Rev. 2 , S11–S17 (1989).
Crew, P. E. et al. Unusual Neisseria species as a cause of infection in patients taking eculizumab. J. Infect. 78 , 113–118 (2019).
Liu, Y., Feinen, B. & Russell, M. W. New concepts in immunity to Neisseria gonorrhoeae : innate responses and suppression of adaptive immunity favor the pathogen, not the host. Front. Microbiol. 2 , 52 (2011).
Boslego, J. W. et al. Efficacy trial of a parenteral gonococcal pilus vaccine in men. Vaccine 9 , 154–162 (1991).
Rotman, E. & Seifert, H. S. The genetics of Neisseria species. Annu. Rev. Genet. 48 , 405–431 (2014).
Bignell, C. & Unemo, M., European STI Guidelines Editorial Board. European guideline on the diagnosis and treatment of gonorrhoea in adults. Int. J. STD & AIDS 24 , 85–92 (2012).
Article Google Scholar
Ghanem, K. G. Clinical manifestations and diagnosis of Neisseria gonorrhoeae infection in adults and adolescents UpToDate.com https://www.uptodate.com/contents/clinical-manifestations-and-diagnosis-of-neisseria-gonorrhoeae-infection-in-adults-and-adolescents/print (2019).
Ison, C. A. Laboratory methods in genitourinary medicine. Methods of diagnosing gonorrhoea. Genitourin. Med. 66 , 453–459 (1990).
Unemo, M. & Ison, C. in Laboratory diagnosis of sexually transmitted infections, including human immunodeficiency virus (eds Unemo, M. et al.) 21–54 (World Health Organization, 2013). This comprehensive chapter describes biological sampling, different methods for laboratory detection and antimicrobial susceptibility testing of N. gonorrhoeae .
Taylor, S. N., DiCarlo, R. P. & Martin, D. H. Comparison of methylene blue/gentian violet stain to Gram's stain for the rapid diagnosis of gonococcal urethritis in men. Sex. Transm. Dis. 38 , 995–996 (2011).
Papp, J. R. S. J., Gaydos, C. A. & Van Der Pol, B. Recommendations for the laboratory-based detection of Chlamydia trachomatis and Neisseria gonorrhoeae — 2014. MMWR Recomm. Rep. 63 , 1–19 (2014).
Google Scholar
Dillon, J. R. Sustainable antimicrobial surveillance programs essential for controlling Neisseria gonorrhoeae superbug. Sex. Transm. Dis. 38 , 899–901 (2011).
Starnino, S. D., J. R. Laboratory manual: identification and antimicrobial susceptibility testing of Neisseria gonorrhoeae 2nd edn Co-ordinating Centre for the Gonococcal Antimicrobial Susceptibility Surveillance Program in Latin America and the Caribbean (2002).
Starnino, S. & Dillon, J. R. in Laboratory diagnosis of sexually transmitted infections, including human immunodeficiency virus (eds Unemo, M. et al.)199–218 (World Health Organization, 2013).
Dillon, J. R., Carballo, M. & Pauze, M. Evaluation of eight methods for identification of pathogenic Neisseria species: Neisseria-Kwik, RIM-N, Gonobio-Test, Minitek, Gonochek II, GonoGen, Phadebact Monoclonal GC OMNI Test, and Syva MicroTrak Test. J. Clin. Microbiol. 26 , 493–497 (1988).
Kellogg, J. A. & Orwig, L. K. Comparison of GonoGen, GonoGen II, and MicroTrak direct fluorescent-antibody test with carbohydrate fermentation for confirmation of culture isolates of Neisseria gonorrhoeae. J. Clin. Microbiol. 33 , 474–476 (1995).
Kulkarni, S., Bala, M. & Risbud, A. Performance of tests for identification of Neisseria gonorrhoeae. Indian J. Med. Res. 141 , 833–835 (2015).
Centers for Disease Control and Prevention. Acid Detection Test http://www.cdc.gov/std/gonorrhea/lab/tests/acid.htm (CDC, 2013).
Buchanan, R., Ball, D., Dolphin, H. & Dave, J. Matrix-assisted laser desorption-ionization time-of-flight mass spectrometry for the identification of Neisseria gonorrhoeae. Clin. Microbiol. Infect. 22 , 815.e815–815.e817 (2016).
Ilina, E. N. et al. Direct bacterial profiling by matrix-assisted laser desorption-ionization time-of-flight mass spectrometry for identification of pathogenic Neisseria . J. Mol. Diagn. 11 , 75–86 (2009).
Morel, F. et al. Use of Andromas and Bruker MALDI-TOF MS in the identification of Neisseria . Eur. J. Clin. Microbiol. Infect. Dis 37 , 2273–2277 (2018).
Schmidt, K. et al. Identification of bacterial pathogens and antimicrobial resistance directly from clinical urines by nanopore-based metagenomic sequencing. J. Antimicrob. Chemother. 72 , 104–114 (2017).
Hughes, G. I. et al . Guidance for the detection of gonorrhoea in England . (Public Health England, London, 2014).
Tabrizi, S. N. et al. Evaluation of six commercial nucleic acid amplification tests for detection of Neisseria gonorrhoeae and other Neisseria species. J. Clin. Microbiol. 49 , 3610–3615 (2011).
Murtagh, M. M. The point-of-care diagnostic landscape for sexually transmitted infections (STIs). WHO https://www.who.int/reproductivehealth/topics/rtis/Diagnostic_Landscape_2018.pdf (2018). This extensive report details point-of-care diagnostic tests for STIs, with special focus on tests in the development pipeline .
Whiley, D. M., Tapsall, J. W. & Sloots, T. P. Nucleic acid amplification testing for Neisseria gonorrhoeae : an ongoing challenge. J. Mol. Diagn. 8 , 3–15 (2006).
Alexander, S., da Silva, Coelho, Manuel, F., Varma, R. & Ison, R. C. Evaluation of strategies for confirming Neisseria gonorrhoeae nucleic acid amplification tests. J. Med. Microbiol. 60 , 909–912 (2011).
Venter, J. M. E. et al. Comparison of an in-house real-time duplex PCR assay with commercial HOLOGIC(R) APTIMA assays for the detection of Neisseria gonorrhoeae and Chlamydia trachomatis in urine and extra-genital specimens. BMC Infect. Dis. 19 , 6 (2019).
United States Food and Drug Administration. Nucleic Acid Based Tests. FDA https://www.fda.gov/medical-devices/vitro-diagnostics/nucleic-acid-based-tests (2019).
Schachter, J., Moncada, J., Liska, S., Shayevich, C. & Klausner, J. D. Nucleic acid amplification tests in the diagnosis of chlamydial and gonococcal infections of the oropharynx and rectum in men who have sex with men. Sex. Transm. Dis. 35 , 637–642 (2008).
Chernesky, M. et al. Head-to-head comparison of second-generation nucleic acid amplification tests for detection of Chlamydia trachomatis and Neisseria gonorrhoeae on urine samples from female subjects and self-collected vaginal swabs. J. Clin. Microbiol. 52 , 2305–2310 (2014).
Jang, D. et al. Comparison of workflow, maintenance, and consumables in the genexpert infinity 80 and panther instruments while testing for chlamydia trachomatis and Neisseria gonorrhoeae . Sex. Transm. Dis. 43 , 377–381 (2016).
World Health Organization. WHO Guidelines for the Treatment of Neisseria gonorrhoeae (WHO, 2016).
Public Health Agency Canada. National Surveillance of Antimicrobial Susceptibilities of Neisseria gonorrhoeae - 2016. (Government of Canada 2018).
Thakur, S. D. & Dillon, J. R. High levels of susceptibility to new and older antibiotics in Neisseria gonorrhoeae isolates from Saskatchewan (2003–15): time to consider point-of-care or molecular testing for precision treatment? Authors' response. J. Antimicrob. Chemother. 73 , 829–830 (2018).
Allan-Blitz, L. T. et al. Implementation of a rapid genotypic assay to promote targeted ciprofloxacin therapy of Neisseria gonorrhoeae in a large health system. Clin. Infect. Dis. 64 , 1268–1270 (2017).
PubMed Google Scholar
Ellis, O. et al. A multisite implementation of a real-time polymerase chain reaction assay to predict ciprofloxacin susceptibility in Neisseria gonorrhoeae . Diagn. Microbiol. Infect. Dis. 94 , 213–217 (2019).
Fifer, H., Saunders, J., Soni, S., Sadiq, S. T. & FitzGerald, M. British Association for Sexual Health and HIV national guideline for the management of infection with Neisseria gonorrhoeae (BASHH, 2019).
Badman, S. G. et al . A diagnostic evaluation of a molecular assay used for testing and treating anorectal chlamydia and gonorrhoea infections at the point-of-care in Papua New Guinea. Clin. Microbiol. Infect . 25 , 623–627 (2018).
Wi, T. E. et al. Diagnosing sexually transmitted infections in resource-constrained settings: challenges and ways forward. J. Int. AIDS Soc. 22 (Suppl. 6), e25343 (2019).
PubMed PubMed Central Google Scholar
Pai, M., Ghiasi., M. & Pai, N. P. Point-of-care diagnostic testing in global health: what is the point? Microbe 10 , 103–107 (2015).
Pai, N. P., Vadnais, C., Denkinger, C., Engel, N. & Pai, M. Point-of-care testing for infectious diseases: diversity, complexity, and barriers in low- and middle-income countries. PLoS Med. 9 , e1001306 (2012).
Peeling, R. W., Holmes, K. K., Mabey, D. & Ronald, A. Rapid tests for sexually transmitted infections (STIs): the way forward. Sex. Transm. Infect. 82 (Suppl. 5), v1–v6 (2006).
Watchirs Smith, L. A. et al. Point-of-care tests for the diagnosis of Neisseria gonorrhoeae infection: a systematic review of operational and performance characteristics. Sex. Transm. Infect. 89 , 320–326 (2013).
Cristillo, A. D. et al. Point-of-care sexually transmitted infection diagnostics: proceedings of the STAR Sexually Transmitted Infection–Clinical Trial Group Programmatic Meeting. Sex. Transm. Dis. 44 , 211–218 (2017).
Herbst de Cortina, S., Bristow, C. C., Joseph Davey, D. & Klausner, J. D. A systematic review of point of care testing for chlamydia trachomatis , Neisseria gonorrhoeae , and trichomonas vaginalis . Infect. Dis. Obstet. Gynecol. 2016 , 4386127 (2016).
Guy, R. J. et al. Performance and operational characteristics of point-of-care tests for the diagnosis of urogenital gonococcal infections. Sex. Transm. Infect. 93 , S16–S21 (2017).
Vickerman, P., Watts, C., Alary, M., Mabey, D. & Peeling, R. W. Sensitivity requirements for the point of care diagnosis of chlamydia trachomatis and Neisseria gonorrhoeae in women. Sex. Transm. Infect. 79 , 363–367 (2003).
Causer, L. M. et al. A field evaluation of a new molecular-based point-of-care test for chlamydia and gonorrhoea in remote aboriginal health services in Australia. Sex. Health 12 , 27–33 (2015).
Garrett, N. et al. Diagnostic accuracy of the Xpert CT/NG and OSOM trichomonas rapid assays for point-of-care STI testing among young women in South Africa: a cross-sectional study. BMJ Open 9 , e026888 (2019).
LeFevre, M. L. Screening for chlamydia and gonorrhea: U.S. preventive services task force recommendation statement. Ann. Intern. Med. 161 , 902–910 (2014).
Workowski, K. A. & Bolan, G. A., Centers for Disease Control and Prevention. Sexually transmitted diseases treatment guidelines, 2015. MMWR. Recomm. Rep. 64 , 1–137 (2015).
Centers for Disease Control and Prevention. Preexposure prophylaxis for the prevention of HIV infection in the United States – 2017 update . (US Public Health Service, 2017).
World Health Organization. Global strategy for the prevention and control of sexually transmitted infections: 2006 – 2015 Breaking the chain of transmission. WHO https://www.who.int/hiv/pub/toolkits/stis_strategy%5B1%5Den.pdf (2007).
Gottlieb, S. L. & Johnston, C. Future prospects for new vaccines against sexually transmitted infections. Curr. Opin. Infect. Dis. 30 , 77–86 (2017).
Jerse, A. E. & Deal, C. D. Vaccine research for gonococcal infections: where are we? Sex. Transm. Infect. 89 , iv63–iv68 (2013).
Zhu, W. et al. Vaccines for gonorrhea: can we rise to the challenge? Front. Microbiol. 2 , 124 (2011).
Edwards, J. L., Jennings, M. P., Apicella, M. A. & Seib, K. L. Is gonococcal disease preventable? The importance of understanding immunity and pathogenesis in vaccine development. Crit. Rev. Microbiol. 42 , 928–941 (2016). This review describes the status of gonococcal vaccine development and, in particular, focuses on the model systems available to evaluate drug and vaccine candidates .
Tramont, E. C. Gonococcal vaccines. Clin. Microbiol. Rev. 2 , S74–S77 (1989).
Paynter, J. et al. Effectiveness of a group B outer membrane vesicle meningococcal vaccine in preventing hospitalization from gonorrhea in New Zealand: a retrospective cohort study. Vaccines 7 , E5 (2019).
Petousis-Harris, H. Impact of meningococcal group B OMV vaccines, beyond their brief. Hum. Vaccin. Immunother. 14 , 1058–1063 (2018).
Petousis-Harris, H. et al. Effectiveness of a group B outer membrane vesicle meningococcal vaccine against gonorrhoea in New Zealand: a retrospective case-control study. Lancet 390 , 1603–1610 (2017). This study provides a first proof-of-principle for vaccine protection against gonorrhoea, owing to cross-protection by the outer membrane vesicle Neisseria meningitidis serogroup B vaccine (MeNZB) .
Hadad, R. et al. Novel meningococcal 4CMenB vaccine antigens — prevalence and polymorphisms of the encoding genes in Neisseria gonorrhoeae . APMIS 120 , 750–760 (2012).
Beernink, P. T. et al. A meningococcal native outer membrane vesicle vaccine with attenuated endotoxin and overexpressed factor h binding protein elicits gonococcal bactericidal antibodies. J. Infect. Dis. 219 , 1130–1137 (2019).
Centers for Disease Control and Prevention. Expedited partner therapy in the management of sexually transmitted diseases. (US Department of Health and Human Services, 2006).
Parran, T. Shadow on the Land: Syphilis . (Reynal & Hitchcock, 1937).
Golden, M. R. et al. Effect of expedited treatment of sex partners on recurrent or persistent gonorrhea or chlamydial infection. N. Engl. J. Med. 352 , 676–685 (2005).
Romanowski, B., Robinson, J. & Wong, T. Canadian Guidelines on Sexually Transmitted Infections - Gonococcal Infections Chapter. Phac-aspc.gc.ca http://www.phac-aspc.gc.ca/std-mts/sti-its/cgsti-ldcits/assets/pdf/section-5-6-eng.pdf (2013).
Australasian Sexual Health Alliance (ASHA). Gonorrhoea. ASHA http://www.sti.guidelines.org.au/sexually-transmissible-infections/gonorrhoea#management (2016).
Japanese Society for Sexually Transmitted Infections. Gonococcal infection. Sexually transmitted infections, diagnosis and treatment guidelines 2011. Jpn J. Sex. Transm. Dis. 22 (Suppl. 1), 52–59 (2011). In Japanese.
Bignell, C. & Fitzgerald, M., Guideline Development Group, British Association for Sexual Health and HIV UK. UK national guideline for the management of gonorrhoea in adults, 2011. Int. J. STD & AIDS 22 , 541–547 (2011).
Boiko, I. et al. Antimicrobial susceptibility of Neisseria gonorrhoeae isolates and treatment of gonorrhoea patients in ternopil and dnipropetrovsk regions of Ukraine, 2013–2018. APMIS 127 , 503–509 (2019).
Unemo, M., Shipitsyna, E. & Domeika, M. Eastern European Sexual and Reproductive Health (EE SRH) Network Antimicrobial Resistance Group. Recommended antimicrobial treatment of uncomplicated gonorrhoea in 2009 in 11 East European countries: implementation of a Neisseria gonorrhoeae antimicrobial susceptibility programme in this region is crucial. Sex. Transm. Infect. 86 , 442–444 (2010).
Leonard, C. A., Schoborg, R. V., Low, N., Unemo, M. & Borel, N. Pathogenic interplay between chlamydia trachomatis and Neisseria gonorrhoeae that influences management and control efforts — more questions than answers? Curr. Clin. Microbiol. Rep. 6 , 182–191 (2019).
Handsfield, H. H., McCutchan, J. A., Corey, L. & Ronald, A. R. Evaluation of new anti-infective drugs for the treatment of uncomplicated gonorrhea in adults and adolescents. infectious diseases society of america and the food and drug administration. Clin. Infect. Dis. 15 (Suppl. 1), S123–S130 (1992).
Hook, E. W. 3rd & Kirkcaldy, R. D. A brief history of evolving diagnostics and therapy for gonorrhea: lessons learned. Clin. Infect. Dis. 67 , 1294–1299 (2018).
Unemo, M. et al. World Health Organization Global Gonococcal Antimicrobial Surveillance Program (WHO GASP): review of new data and evidence to inform international collaborative actions and research efforts. Sex. Health 16 , 412–425 (2019). This paper reports the WHO GASP data from 2015 to 2016, confirmed gonorrhoea treatment failures with recommended therapy and international collaborative actions and research efforts essential for the effective management and control of gonorrhoea .
Cole, M. J. et al. Is the tide turning again for cephalosporin resistance in Neisseria gonorrhoeae in Europe? Results from the 2013 European surveillance. BMC Infect. Dis. 15 , 321 (2015).
Day, M. J. et al. Stably high azithromycin resistance and decreasing ceftriaxone susceptibility in Neisseria gonorrhoeae in 25 European countries, 2016. BMC Infect. Dis. 18 , 609 (2018).
Harris, S. R. et al. Public health surveillance of multidrug-resistant clones of Neisseria gonorrhoeae in Europe: a genomic survey. Lancet Infect. Dis. 18 , 758–768 (2018). This genomics paper provides the first use of joint analysis of WGS and epidemiological data in an international surveillance programme for STIs and a framework for genomic surveillance of gonococci through standardized sampling, use of WGS, and a shared information architecture for interpretation and dissemination by use of open-access software .
Kirkcaldy, R. D. et al. Neisseria gonorrhoeae antimicrobial susceptibility surveillance — the gonococcal isolate surveillance project, 27 sites, United States, 2014. MMWR 65 , 1–19 (2016).
Kirkcaldy, R. D., Kidd, S., Weinstock, H. S., Papp, J. R. & Bolan, G. A. Trends in antimicrobial resistance in Neisseria gonorrhoeae in the USA: the Gonococcal Isolate Surveillance Project (GISP), January 2006–June 2012. Sex. Transm. Infect. 89 , iv5–iv10 (2013).
Ford, J. V. et al. The need to promote sexual health in America: a new vision for public health action. Sex. Transm. Dis. 44 , 579–585 (2017).
Reed, J. L. et al. Adolescent patient preferences surrounding partner notification and treatment for sexually transmitted infections. Acad. Emerg. Med. 22 , 61–66 (2015).
Goffman, E. Stigma: notes on the management of spoiled identity (Aronson, J., 1974).
Fortenberry, J. D. et al. Relationships of stigma and shame to gonorrhea and HIV screening. Am. J. Public Health 92 , 378–381 (2002).
Lichtenstein, B. Stigma as a barrier to treatment of sexually transmitted infection in the American deep south: issues of race, gender and poverty. Soc. Sci. Med. 57 , 2435–2445 (2003).
Tsadik, M., Berhane, Y., Worku, A. & Terefe, W. The magnitude of, and factors associated with, loss to follow-up among patients treated for sexually transmitted infections: a multilevel analysis. BMJ Open 7 , e016864 (2017).
Tshokey, T. et al. Antibiotic resistance in Neisseria gonorrhoea and treatment outcomes of gonococcal urethritis suspected patients in two large hospitals in Bhutan, 2015. PLoS One 13 , e0201721 (2018).
Schwartz, R. M. et al. Coping with a diagnosis of C. trachomatis or N. gonorrhoeae : psychosocial and behavioral correlates. J. Health Psychol. 13 , 921–929 (2008).
Wong, J. P. H., Chan, K. B. K., Bio-Doku, R. & Mcwatt, S. Risk discourse and sexual stigma: barriers to STI testing, treatment and care among young heterosexual women in disadvantaged neighbourhoods in Toronto. Can. J. Hum. Sex. 21 , 74–89 (2012).
Morris, J. L. et al. Sexually transmitted infection related stigma and shame among African American male youth: implications for testing practices, partner notification, and treatment. AIDS Patient Care STDS 28 , 499–506 (2014).
Crenshaw, K. Mapping the margins: intersectionality, identity politics, and violence against women of color. Stanf. Law Rev. 43 , 1241–1299 (1991).
Unemo, M. et al. Sexually transmitted infections: challenges ahead. Lancet Infect. Dis. 17 , e235–e279 (2017). This very extensive Commission discusses the current key challenges facing the field of STIs and outlines new approaches to improve the clinical management of STIs and public health .
Carlton, T. O. & Mayes, S. M. Gonorrhea: not a ‘second-class’ disease. Health Soc. Work. 7 , 301–313 (1982).
Wu, D., Hawkes, S. & Buse, K. Prevention of mother-to-child transmission of syphilis and HIV in China: what drives political prioritization and what can this tell us about promoting dual elimination? Int. J. Gynaecol. Obstet. 130 , S32–S36 (2015).
Cook, J. E., Purdie-Vaughns, V., Meyer, I. H. & Busch, J. T. A. Intervening within and across levels: a multilevel approach to stigma and public health. Soc. Sci. Med. 103 , 101–109 (2014).
Demczuk, W. et al. Whole-genome phylogenomic heterogeneity of Neisseria gonorrhoeae isolates with decreased cephalosporin susceptibility collected in Canada between 1989 and 2013. J. Clin. Microbiol. 53 , 191–200 (2015).
Article PubMed CAS Google Scholar
Demczuk, W. et al. Genomic epidemiology and molecular resistance mechanisms of azithromycin-resistant Neisseria gonorrhoeae in Canada from 1997 to 2014. J. Clin. Microbiol. 54 , 1304–1313 (2016).
Grad, Y. H. et al. Genomic epidemiology of Neisseria gonorrhoeae with reduced susceptibility to cefixime in the USA: a retrospective observational study. Lancet Infect. Dis. 14 , 220–226 (2014).
Grad, Y. H. et al. Genomic epidemiology of gonococcal resistance to extended-spectrum cephalosporins, macrolides, and fluoroquinolones in the United States, 2000–2013. J. Infect. Dis. 214 , 1579–1587 (2016).
Jacobsson, S. et al. WGS analysis and molecular resistance mechanisms of azithromycin-resistant (MIC >2 mg/L) Neisseria gonorrhoeae isolates in Europe from 2009 to 2014. J. Antimicrob. Chemother. 71 , 3109–3116 (2016).
De Silva, D. et al. Whole-genome sequencing to determine transmission of Neisseria gonorrhoeae : an observational study. Lancet Infect. Dis. 16 , 1295–1303 (2016).
Ezewudo, M. N. et al. Population structure of Neisseria gonorrhoeae based on whole genome data and its relationship with antibiotic resistance. PeerJ. 3 , e806 (2015).
Ryan, L. et al. Antimicrobial resistance and molecular epidemiology using whole-genome sequencing of Neisseria gonorrhoeae in Ireland, 2014–2016: focus on extended-spectrum cephalosporins and azithromycin. Eur. J. Clin. Microbiol. Infect. Dis. 37 , 1661–1672 (2018).
Eyre, D. W. et al. WGS to predict antibiotic MICs for Neisseria gonorrhoeae . J. Antimicrob. Chemother. 72 , 1937–1947 (2017). This genomics paper provides strong evidence that WGS can relatively successfully predict MICs of antimicrobials and AMR in N. gonorrhoeae .
Golparian, D. et al. Antimicrobial resistance prediction and phylogenetic analysis of Neisseria gonorrhoeae isolates using the Oxford Nanopore MinION sequencer. Sci. Rep. 8 , 17596 (2018).
Eyre, D. W., Golparian, D. & Unemo, M. Prediction of minimum inhibitory concentrations of antimicrobials for Neisseria gonorrhoeae using whole-genome sequencing. Methods Mol. Biol. 1997 , 59–76 (2019).
Unemo, M. & Althaus, C. L. Fitness cost and benefit of antimicrobial resistance in Neisseria gonorrhoeae : multidisciplinary approaches are needed. PLoS Med. 14 , e1002423 (2017).
Fingerhuth, S. M., Low, N., Bonhoeffer, S. & Althaus, C. L. Detection of antibiotic resistance is essential for gonorrhoea point-of-care testing: a mathematical modelling study. BMC Med. 15 , 142 (2017).
Jacobsson, S. et al. WHO laboratory validation of Xpert((R)) CT/NG and Xpert((R)) TV on the GeneXpert system verifies high performances. APMIS. 126 , 907–912 (2018).
Nudel, K. et al. Transcriptome analysis of Neisseria gonorrhoeae during natural infection reveals differential expression of antibiotic resistance determinants between men and women. mSphere 3 , e00312–e00318 (2018).
Zielke, R. A. et al. Proteomics-driven antigen discovery for development of vaccines against gonorrhea. Mol. Cell Proteomics 15 , 2338–2355 (2016).
El-Rami, F. E., Zielke, R. A., Wi, T., Sikora, A. E. & Unemo, M. Quantitative proteomics of the 2016 WHO Neisseria gonorrhoeae reference strains surveys vaccine candidates and antimicrobial resistance determinants. Mol. Cell Proteomics 18 , 127–150 (2019).
Unemo, M. & Sikora, A. E. Infection: proof of principle for effectiveness of a gonorrhoea vaccine. Nat. Rev. Urol. 14 , 643–644 (2017).
Moreau, M. R., Massari, P. & Genco, C. A. The ironclad truth: how in vivo transcriptomics and in vitro mechanistic studies shape our understanding of Neisseria gonorrhoeae gene regulation during mucosal infection. Pathog. Dis . 75 , https://doi.org/10.1093/femspd/ftx057 (2017).
Jerse, A. E. et al. Estradiol-treated female mice as surrogate hosts for Neisseria gonorrhoeae genital tract infections. Front. Microbiol. 2 , 107 (2011).
Sintsova, A. et al . Selection for CEACAM receptor-specific binding phenotype during Neisseria gonorrhoeae infection of the human genital tract. Infect. Immun. 83 , 1372–1383 (2015).
Lujan, E., Pajon, R. & Granoff, D. M. Impaired immunogenicity of meningococcal neisserial surface protein A in human complement factor H transgenic mice. Infect. Immun. 84 , 452–458 (2016).
Low, N. & Unemo, M. Molecular tests for the detection of antimicrobial resistant Neisseria gonorrhoeae : when, where, and how to use? Curr. Opin. Infect. Dis. 29 , 45–51 (2016).
Dona, V., Low, N., Golparian, D. & Unemo, M. Recent advances in the development and use of molecular tests to predict antimicrobial resistance in Neisseria gonorrhoeae . Expert Rev. Mol. Diagn. 17 , 845–859 (2017).
Sadiq, S. T., Mazzaferri, F. & Unemo, M. Rapid accurate point-of-care tests combining diagnostics and antimicrobial resistance prediction for Neisseria gonorrhoeae and mycoplasma genitalium. Sex. Transm. Infect. 93 , S65–S68 (2017).
Goire, N. et al. Molecular approaches to enhance surveillance of gonococcal antimicrobial resistance. Nat. Rev. Microbiol. 12 , 223–229 (2014).
Basarab, G. S. et al. Responding to the challenge of untreatable gonorrhea: ETX0914, a first-in-class agent with a distinct mechanism-of-action against bacterial type II topoisomerases. Sci. Rep. 5 , 11827 (2015).
Foerster, S. et al. Genetic resistance determinants, in vitro time-kill curve analysis and pharmacodynamic functions for the novel topoisomerase II inhibitor ETX0914 (AZD0914) in Neisseria gonorrhoeae . Front. Microbiol. 6 , 1377 (2015).
Jacobsson, S. et al. High in vitro activity of the novel spiropyrimidinetrione AZD0914, a DNA gyrase inhibitor, against multidrug-resistant Neisseria gonorrhoeae isolates suggests a new effective option for oral treatment of gonorrhea. Antimicrob. Agents Chemother. 58 , 5585–5588 (2014).
Taylor, S. N. et al. Single-dose zoliflodacin (ETX0914) for treatment of urogenital gonorrhea. N. Engl. J. Med. 379 , 1835–1845 (2018).
Foerster, S. et al . In vitro antimicrobial combination testing and evolution of resistance to the first-in-class spiropyrimidinetrione zoliflodacin combined with six therapeutically relevant antimicrobials for Neisseria gonorrhoeae . J. Antimicrob. Chemother . https://doi.org/10.1093/jac/dkz376 (2019)
Jacobsson, S., Golparian, D., Scangarella-Oman, N. & Unemo, M. In vitro activity of the novel triazaacenaphthylene gepotidacin (GSK2140944) against MDR Neisseria gonorrhoeae . J. Antimicrob. Chemother. 73 , 2072–2077 (2018).
Scangarella-Oman, N. E. et al . Microbiological analysis from a phase 2 randomized study in adults evaluating single oral doses of gepotidacin in the treatment of uncomplicated urogenital gonorrhea caused by Neisseria gonorrhoeae . Antimicrob. Agents Chemother . 62 , e01221–18 (2018).
Taylor, S. N. et al. Gepotidacin for the treatment of uncomplicated urogenital gonorrhea: a phase 2, randomized, dose-ranging, single-oral dose evaluation. Clin. Infect. Dis. 67 , 504–512 (2018).
Jacobsson, S., Paukner, S., Golparian, D., Jensen, J. S. & Unemo, M. In vitro activity of the novel pleuromutilin lefamulin (bc-3781) and effect of efflux pump inactivation on multidrug-resistant and extensively drug-resistant Neisseria gonorrhoeae . Antimicrob. Agents Chemother. 61 , e01497–17 (2017).
Paukner, S., Gruss, A. & Jensen, J. S. In vitro activity of lefamulin against sexually transmitted bacterial pathogens. Antimicrob. Agents Chemother. 62 , e02380–17 (2018).
Jacobsson, S., Mason, C., Khan, N., Meo, P. & Unemo, M. In vitro activity of the novel oral antimicrobial SMT-571, with a new mechanism of action, against MDR and XDR Neisseria gonorrhoeae: future treatment option for gonorrhoea? J. Antimicrob. Chemother. 74 , 1591–1594 (2019).
Kong, F. Y. S., Horner, P., Unemo, M. & Hocking, J. S. Pharmacokinetic considerations regarding the treatment of bacterial sexually transmitted infections with azithromycin: a review. J. Antimicrob. Chemother. 74 , 1157–1166 (2019). This paper provides a detailed overview of the pharmacokinetics of antimicrobials used to treat STIs and how factors related to the drug, human and organism can affect treatment outcomes .
Lenz, J. D. & Dillard, J. P. Pathogenesis of Neisseria gonorrhoeae and the host defense in ascending infections of human fallopian tube. Front. Immunol. 9 , 2710 (2018).
Lucas, C. T., Chandler, F. Jr., Martin, J. E. Jr & Schmale, J. D. Transfer of gonococcal urethritis from man to chimpanzee. An animal model for gonorrhea. JAMA 216 , 1612–1614 (1971).
Cohen, M. S. & Cannon, J. G. Human experimentation with Neisseria gonorrhoeae : progress and goals. J. Infect. Dis. 179 (Suppl. 2), S375–S379 (1999).
Chow, E. P. et al. Antiseptic mouthwash against pharyngeal Neisseria gonorrhoeae : a randomised controlled trial and an in vitro study. Sex. Transm. Infect. 93 , 88–93 (2017).
Liu, Y. et al. Experimental vaccine induces Th1-driven immune responses and resistance to Neisseria gonorrhoeae infection in a murine model. Mucosal Immunol. 10 , 1594–1608 (2017).
Kenyon, C., Buyze, J., Spiteri, G., Cole, M. J. & Unemo, M. Population-level antimicrobial consumption is associated with decreased antimicrobial susceptibility in Neisseria gonorrhoeae in 24 European countries: an ecological analysis. J. Infect. Dis . https://doi.org/10.1093/infdis/jiz153 (2019).
Tomberg, J. et al. Alanine 501 mutations in penicillin-binding protein 2 from Neisseria gonorrhoeae : structure, mechanism, and effects on cephalosporin resistance and biological fitness. Biochemistry 56 , 1140–1150 (2017).
Tomberg, J., Unemo, M., Davies, C. & Nicholas, R. A. Molecular and structural analysis of mosaic variants of penicillin-binding protein 2 conferring decreased susceptibility to expanded-spectrum cephalosporins in Neisseria gonorrhoeae : role of epistatic mutations. Biochemistry 49 , 8062–8070 (2010).
Tomberg, J., Unemo, M., Ohnishi, M., Davies, C. & Nicholas, R. A. Identification of amino acids conferring high-level resistance to expanded-spectrum cephalosporins in the penA gene from Neisseria gonorrhoeae strain H041. Antimicrob. Agents Chemother. 57 , 3029–3036 (2013).
Lee, H. et al. Emergence of decreased susceptibility and resistance to extended-spectrum cephalosporins in Neisseria gonorrhoeae in Korea. J. Antimicrob. Chemother. 70 , 2536–2542 (2015).
Olsen, B. et al. Antimicrobial susceptibility and genetic characteristics of Neisseria gonorrhoeae isolates from Vietnam, 2011. BMC Infect. Dis. 13 , 40 (2013).
Whiley, D. M. et al. Reduced susceptibility to ceftriaxone in Neisseria gonorrhoeae is associated with mutations G542S, P551S and P551L in the gonococcal penicillin-binding protein 2. J. Antimicrob. Chemother. 65 , 1615–1618 (2010).
Ohnishi, M. 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. 55 , 3538–3545 (2011). This paper describes the identification and verification of the first global extensively drug-resistant and high-level ceftriaxone-resistant gonococcal strain that caused a ceftriaxone treatment failure in Japan .
Camara, J. et al. Molecular characterization of two high-level ceftriaxone-resistant Neisseria gonorrhoeae isolates detected in Catalonia, Spain. J. Antimicrob. Chemother. 67 , 1858–1860 (2012).
Unemo, M. 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. 56 , 1273–1280 (2012).
Gianecini, R., Oviedo, C., Stafforini, G. & Galarza, P. Neisseria gonorrhoeae resistant to ceftriaxone and cefixime, Argentina. Emerg. Infect. Dis. 22 , 1139–1141 (2016).
Deguchi, T. et al. New clinical strain of Neisseria gonorrhoeae with decreased susceptibility to ceftriaxone, Japan. Emerg. Infect. Dis. 22 , 142–144 (2016).
Nakayama, S. et al. New ceftriaxone- and multidrug-resistant Neisseria gonorrhoeae strain with a novel mosaic pena gene isolated in Japan. Antimicrob. Agents Chemother. 60 , 4339–4341 (2016).
Lahra, M. M. et al . Cooperative recognition of internationally disseminated ceftriaxone-resistant Neisseria gonorrhoeae strain. Emerg. Infect. Dis . 24 , https://doi.org/10.3201/eid2404.171873 (2018).
Lefebvre, B. et al . Ceftriaxone-resistant Neisseria gonorrhoeae , Canada, 2017. Emerg. Infect. Dis . 24 , https://doi.org/10.3201/eid2402.171756 (2018).
Article CAS PubMed Central Google Scholar
Terkelsen, D. et al . Multidrug-resistant Neisseria gonorrhoeae infection with ceftriaxone resistance and intermediate resistance to azithromycin, Denmark, 2017. Euro Surveill 22 , https://doi.org/10.2807/1560-7917.ES.2017.22.42.17-00659 (2017).
Poncin, T. et al . Multidrug-resistant Neisseria gonorrhoeae failing treatment with ceftriaxone and doxycycline in France, November 2017. Euro Surveill 23 , https://doi.org/10.2807/1560-7917.ES.2018.23.21.1800264 (2018).
Golparian, D. et al . Multidrug-resistant Neisseria gonorrhoeae isolate, belonging to the internationally spreading Japanese FC428 clone, with ceftriaxone resistance and intermediate resistance to azithromycin, Ireland, August 2018. Euro Surveill 23 , https://doi.org/10.2807/1560-7917.ES.2018.23.47.1800617 (2018).
Eyre, D. W. et al . Detection in the United Kingdom of the Neisseria gonorrhoeae FC428 clone, with ceftriaxone resistance and intermediate resistance to azithromycin, October to December 2018. Euro Surveill 24 , https://doi.org/10.2807/1560-7917.ES.2019.24.10.1900147 (2019).
Eyre, D. W. et al . Gonorrhoea treatment failure caused by a Neisseria gonorrhoeae strain with combined ceftriaxone and high-level azithromycin resistance, England, February 2018. Euro Surveill 23 , https://doi.org/10.2807/1560-7917.ES.2018.23.27.1800323 (2018). This paper describes the identification of the first global gonococcal strain with combined ceftriaxone and high-level azithromycin resistance that caused a ceftriaxone treatment failure in the UK .
Whiley, D. M., Jennison, A., Pearson, J. & Lahra, M. M. Genetic characterisation of Neisseria gonorrhoeae resistant to both ceftriaxone and azithromycin. Lancet Infect. Dis. 18 , 717–718 (2018).
Jennison, A. V. et al . Genetic relatedness of ceftriaxone-resistant and high-level azithromycin-resistant Neisseria gonorrhoeae cases, United Kingdom and Australia, February to April 2018. Euro Surveill 24 , https://doi.org/10.2807/1560-7917.ES.2019.24.8.1900118 (2019).
Ko, K. K. K. et al. First case of ceftriaxone-resistant multidrug-resistant Neisseria gonorrhoeae in Singapore. Antimicrob. Agents Chemother 63 , e06224-18 (2019).
Lee, K. et al. Clonal expansion and spread of the ceftriaxone-resistant Neisseria gonorrhoeae strain FC428, identified in Japan in 2015, and closely related isolates. J. Antimicrob. Chemother 74 , 1812–1819 (2019).
Fifer, H. et al. Failure of dual antimicrobial therapy in treatment of gonorrhea. N. Engl. J. Med. 374 , 2504–2506 (2016). This study reports on the first global failure of dual antimicrobial therapy (ceftriaxone plus azithromycin) in the treatment of gonorrhoea .
Chen, S. C., Han, Y., Yuan, L. F., Zhu, X. Y. & Yin, Y. P. Identification of internationally disseminated ceftriaxone-resistant Neisseria gonorrhoeae strain FC428, China. Emerg. Infect. Dis. 25 , 1427–1429 (2019).
Poncin, T. et al . Two cases of multidrug-resistant Neisseria gonorrhoeae related to travel in south-eastern Asia, France, June 2019. Euro Surveill 24 , https://doi.org/10.2807/1560-7917.ES.2019.24.36.1900528 (2019).
Morse, S. A. The biology of the gonococcus. CRC Crit. Rev. Microbiol. 7 , 93–189 (1978).
Tonjum, T. & Koomey, M. The pilus colonization factor of pathogenic neisserial species: organelle biogenesis and structure/function relationships — a review. Gene 192 , 155–163 (1997).
Maier, B., Potter, L., So, M., Seifert, H. S. & Sheetz, M. P. Single pilus motor forces exceed 100 pN. Proc. Natl Acad. Sci. USA 99 , 16012–16017 (2002).
Stern, A., Brown, M., Nickel, P. & Meyer, T. F. Opacity genes in Neisseria gonorrhoeae : control of phase and antigenic variation. Cell 47 , 61–71 (1986).
James, J. F. & Swanson, J. Studies on gonococcus infection. XIII. Occurrence color/opacity colonial variants in clinical cultures. Infect. Immun. 19 , 332–340 (1978).
Jerse, A. E. et al. Multiple gonococcal opacity proteins are expressed during experimental urethral infection in the male. J. Exp. Med. 179 , 911–920 (1994).
Rice, P. A., Vayo, H. E., Tam, M. R. & Blake, M. S. Immunoglobulin G antibodies directed against protein III block killing of serum-resistant Neisseria gonorrhoeae by immune serum. J. Exp. Med. 164 , 1735–1748 (1986).
Mandrell, R. E. et al. In vitro and in vivo modification of Neisseria gonorrhoeae lipooligosaccharide epitope structure by sialylation. J. Exp. Med. 171 , 1649–1664 (1990).
Gaydos, C. A. et al. Performance of the Abbott RealTime CT/NG for detection of Chlamydia trachomatis and Neisseria gonorrhoeae . J. Clin. Microbiol. 48 , 3236–3243 (2010).
Levett, P. N. et al. Evaluation of three automated nucleic acid amplification systems for detection of Chlamydia trachomatis and Neisseria gonorrhoeae in first-void urine specimens. J. Clin. Microbiol. 46 , 2109–2111 (2008).
Gaydos, C. A. et al. Performance of the cepheid CT/NG xpert rapid PCR test for detection of Chlamydia trachomatis and Neisseria gonorrhoeae . J. Clin. Microbiol. 51 , 1666–1672 (2013).
Tabrizi, S. N. et al. Analytical evaluation of GeneXpert CT/NG, the first genetic point-of-care assay for simultaneous detection of Neisseria gonorrhoeae and Chlamydia trachomatis . J. Clin. Microbiol. 51 , 1945–1947 (2013).
Bromhead, C., Miller, A., Jones, M. & Whiley, D. Comparison of the cobas 4800 CT/NG test with culture for detecting Neisseria gonorrhoeae in genital and nongenital specimens in a low-prevalence population in New Zealand. J. Clin. Microbiol. 51 , 1505–1509 (2013).
Rockett, R. et al. Evaluation of the cobas 4800 CT/NG test for detecting Chlamydia trachomatis and Neisseria gonorrhoeae . Sex. Transm. Infect. 86 , 470–473 (2010).
Van Der Pol, B., Williams, J. A., Fuller, D., Taylor, S. N. & Hook, E. W. 3rd Combined testing for chlamydia, gonorrhea, and trichomonas by use of the BD Max CT/GC/TV assay with genitourinary specimen types. J. Clin. Microbiol. 55 , 155–164 (2017).
Masek, B. J. et al. Performance of three nucleic acid amplification tests for detection of chlamydia trachomatis and Neisseria gonorrhoeae by use of self-collected vaginal swabs obtained via an internet-based screening program. J. Clin. Microbiol. 47 , 1663–1667 (2009).
Moncada, J., Schachter, J., Liska, S., Shayevich, C. & Klausner, J. D. Evaluation of self-collected glans and rectal swabs from men who have sex with men for detection of Chlamydia trachomatis and Neisseria gonorrhoeae by use of nucleic acid amplification tests. J. Clin. Microbiol. 47 , 1657–1662 (2009).
Golparian, D., Tabrizi, S. N. & Unemo, M. Analytical specificity and sensitivity of the APTIMA Combo 2 and APTIMA GC assays for detection of commensal Neisseria species and Neisseria gonorrhoeae on the gen-probe panther instrument. Sex. Transm. Dis. 40 , 175–178 (2013).
Download references
Acknowledgements
The authors are grateful to S. Jacobsson (Örebro University Hospital and Örebro University) and S. Perera and N. Parmar (University of Saskatchewan) for technical assistance with preparing this manuscript.
Reviewer information
Nature Reviews Disease Primers thanks G. Hughes, S. Sood and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Author information
Authors and affiliations.
World Health Organization Collaborating Centre for Gonorrhoea and other Sexually Transmitted Infections, Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
Magnus Unemo
National Reference Laboratory for Sexually Transmitted Infections, Department of Laboratory Medicine, Microbiology, Örebro University Hospital, Örebro, Sweden
Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
H Steven Seifert
Departments of Medicine, Epidemiology and Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
Edward W. Hook III
Institute for Global Health, University College London, London, UK
Sarah Hawkes
Skin and Genitourinary Medicine Clinic, Harare, Zimbabwe
Francis Ndowa
Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
Jo-Anne R. Dillon
Vaccine and Infectious Disease Organization — International Vaccine Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
You can also search for this author in PubMed Google Scholar
Contributions
Introduction (M.U.); Epidemiology (F.N.); Mechanisms/pathophysiology (H S.S.); Diagnosis, screening and prevention (J.-A.R.D.); Management (E.W.H.III); Quality of life (S.H.); Outlook (M.U.); Overview of Primer (M.U.).
Corresponding author
Correspondence to Magnus Unemo .
Ethics declarations
Competing interests.
All authors declare no competing interests.
Additional information
Publisher’s note.
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Related links
World Bank Income Classification: https://databank.worldbank.org/reports.aspx?source=2&series=NY.GNP.PCAP.CD&country=
Rights and permissions
Reprints and permissions
About this article
Cite this article.
Unemo, M., Seifert, H.S., Hook, E.W. et al. Gonorrhoea. Nat Rev Dis Primers 5 , 79 (2019). https://doi.org/10.1038/s41572-019-0128-6
Download citation
Accepted : 09 October 2019
Published : 21 November 2019
DOI : https://doi.org/10.1038/s41572-019-0128-6
Share this article
Anyone you share the following link with will be able to read this content:
Sorry, a shareable link is not currently available for this article.
Provided by the Springer Nature SharedIt content-sharing initiative
This article is cited by
Neisseria gonorrhoeae antimicrobial resistance patterns and associated risk factors in women of childbearing potential in northwestern ethiopia.
- Engdawork Demissie
- Azanaw Amare
- Mucheye Gizachew
BMC Women's Health (2024)
A role for the ATP-dependent DNA ligase lig E of Neisseria gonorrhoeae in biofilm formation
- Adele Williamson
BMC Microbiology (2024)
Epidemiology of gonorrhea in countries of the Middle East and North Africa: systematic review, meta analyses, and meta regressions
- Hiam Chemaitelly
- Manale Harfouche
- Laith J. Abu-Raddad
BMC Global and Public Health (2024)
Policy, practice, and prediction: model-based approaches to evaluating N. gonorrhoeae antibiotic susceptibility test uptake in Australia
- Phu Cong Do
- Yibeltal Alemu Assefa
- Simon Andrew Reid
BMC Infectious Diseases (2024)
Hormonal steroids induce multidrug resistance and stress response genes in Neisseria gonorrhoeae by binding to MtrR
- Grace M. Hooks
- Julio C. Ayala
- Richard G. Brennan
Nature Communications (2024)
Quick links
- Explore articles by subject
- Guide to authors
- Editorial policies
Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.
An official website of the United States government
Official websites use .gov A .gov website belongs to an official government organization in the United States.
Secure .gov websites use HTTPS A lock ( Lock Locked padlock icon ) or https:// means you've safely connected to the .gov website. Share sensitive information only on official, secure websites.
- Publications
- Account settings
- Advanced Search
- Journal List
Gonorrhea - an evolving disease of the new millennium
Stuart a hill, thao l masters, jenny wachter.
- Author information
- Article notes
- Copyright and License information
* E-mail: [email protected]
Conflict of interest: The authors declare no conflict of interest.
Please cite this article as: Stuart A. Hill, Thao L. Masters and Jenny Wachter (2016). Gonorrhea - an evolving disease of the new millennium. Microbial Cell 3(9): 371-389. doi: 10.15698/mic2016.09.524
Received 2015 Oct 9; Accepted 2016 Jan 30; Collection date 2016 Sep 5.
This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Etiology, transmission and protection: Neisseria gonorrhoeae (the gonococcus) is the etiological agent for the strictly human sexually transmitted disease gonorrhea. Infections lead to limited immunity, therefore individuals can become repeatedly infected. Pathology/symptomatology: Gonorrhea is generally a non-complicated mucosal infection with a pustular discharge. More severe sequellae include salpingitis and pelvic inflammatory disease which may lead to sterility and/or ectopic pregnancy. Occasionally, the organism can disseminate as a bloodstream infection. Epidemiology, incidence and prevalence: Gonorrhea is a global disease infecting approximately 60 million people annually. In the United States there are approximately 300, 000 cases each year, with an incidence of approximately 100 cases per 100,000 population. Treatment and curability: Gonorrhea is susceptible to an array of antibiotics. Antibiotic resistance is becoming a major problem and there are fears that the gonococcus will become the next “superbug” as the antibiotic arsenal diminishes. Currently, third generation extended-spectrum cephalosporins are being prescribed. Molecular mechanisms of infection: Gonococci elaborate numerous strategies to thwart the immune system. The organism engages in extensive phase (on/off switching) and antigenic variation of several surface antigens. The organism expresses IgA protease which cleaves mucosal antibody. The organism can become serum resistant due to its ability to sialylate lipooligosaccharide in conjunction with its ability to subvert complement activation. The gonococcus can survive within neutrophils as well as in several other lymphocytic cells. The organism manipulates the immune response such that no immune memory is generated which leads to a lack of protective immunity.
Keywords: pathogenesis, antigenic variation, immune manipulation, antibiotic resistance, panmictic
INTRODUCTION
Neisseria gonorrhoeae (the gonococcus) is a Gram-negative diplococcus, an obligate human pathogen, and the etiologic agent of the sexually transmitted disease, gonorrhea. The gonococcus infects a diverse array of mucosal surfaces, some of which include the urethra, the endocervix, the pharynx, conjunctiva and the rectum 1 . In 2013, the Centers for Disease Control and Prevention (CDC) reported that there were 333,004 new cases of gonorrhea in the United States, with an incidence of 106.1 cases per 100,000 population 2 . Worldwide, 106.1 million people are infected by N. gonorrhoeae annually 3 . In most cases, the disease is a noncomplicated mucosal infection. However, in a few patients, generally with women, more serious sequelae can occur and include salpingitis (acute inflammation of the fallopian tubes), pelvic inflammatory disease (PID; an infection in the upper part of the female reproductive system), or, in rare cases, as a bacteremic infection 4 . If left untreated, these more serious complications can result in sterility, ectopic pregnancy, septic arthritis, and occasionally death. Approximately 3% of women presenting with a urogenital infection develop the most severe forms of the disease 5 . However, the occurrence of PID has significantly decreased over time 6 , 7 , 8 , with an estimated 40,000 cases of infertility in women annually 9 . Dissemination rarely occurs, but when the bacteria do cross the endothelium, they can spread to other locations in the body. Currently, a more worrying trend has emerged, in that, there now appears to be an increased risk for HIV infection in patients that are also infected with N. gonorrhoeae 10 .
Gonorrhea the disease was initially described approximately 3,500 years ago, but it was not until 1879 that Albert Neisser determined the etiologic agent of the disease 11 . The Neisseriae are usually regarded as microaerophilic organisms. However, under the appropriate conditions, they are capable of anaerobic growth 12 . In vitro cultivation of this fastidious organism has always been problematic and it was not until the development of an improved Thayer-Martin medium that early epidemiological studies could be undertaken. Subsequently, other commercial growth mediums have since been developed which has allowed for a greater understanding of the disease process.
VIRULENCE FACTORS OF N. GONORRHOEAE
Like many Gram-negative bacterial pathogens, N. gonorrhoeae possesses a wide range of virulence determinants, which include the elaboration of pili, Opa protein expression, lipooligosaccharide expression (LOS), Por protein expression and IgA1 protease production that facilitates adaptation within the host.
Type IV pili (Tfp)
Considerable attention was paid to pili stemming from the observations of Kellogg and coworkers 12 , 13 that virulent (T1, T2 organisms) and avirulent (T3, T4 organisms) strains could be differentiated on the basis of colony morphology following growth on solid medium. Subsequently, it was established that all freshly isolated gonococci possessed thin hair-like appendages (pili) which were predominantly composed of protein initially called pilin but subsequently renamed PilE 14 . The elaboration of pili is a critical requirement for infection as this structure plays a primary role in attaching to human mucosal epithelial cells 15 , fallopian tube mucosa 16 , 17 , vaginal epithelial cells 16 , 18 as well as to human polymorphonuclear leukocytes (PMN’s; neutrophils) 19 , 20 . Due to their prominent surface location, pili were initially thought to be an ideal vaccine candidate as pilus-specific antibodies were observed in genital secretions 18 . However, two prominent vaccine trials failed, with evidence indicating that pilus protein(s) underwent antigenic variation 21 .
Gonococcal pili are categorized as Type IV pili, as the PilE polypeptide is initially synthesized with a short (7 amino acid) N-terminal leader peptide, which is then endo-proteolytically cleaved 22 . The mature PilE polypeptide is then assembled at the inner membrane into an emerging pilus organelle with the PilE polypeptides being stacked in an α-helical array 23 . The PilE polypeptide consists of three functional domains based on sequence characteristics 24 . The N-terminal domain is highly conserved and is strongly hydrophobic, with this region of the protein comprising the core of the pilus structure 23 . The central part of the PilE monomer is partially conserved and structurally aligned as a β-pleated sheet. As the C-terminal domain is hydrophilic, this segment of the protein is exposed to the external environment 23 and undergoes antigenic variation which allows the bacteria to avoid recognition by the human host’s immune cells (reviewed 25 , 26 ).
Assembly of the pilus structure is complicated and involves other proteins besides PilE (e.g., the pilus tip-located adhesion, PilC) 27 as well as other minor pilus components PilD, PilF, PilG, PilT, PilP and PilQ 28 . During pilus biogenesis, and prior to assembly, the leader peptide is removed from PilE by the PilD peptidase 23 . The N-terminal domain then facilitates translocation across the cytoplasmic membrane allowing PilE subunits to be polymerized at the inner membrane 29 , 30 . As the pilus structure is assembled, it is extruded to the exterior of the outer membrane using the PilQ pore forming complex 29 , 30 , 31 . PilC is a minor protein located at the tip of pilus as well as being present at its base. The pilC gene exists as 2 homologous, but non-identical copies, pilC1 and pilC2 in most gonococcal strains, with only the pilC2 gene being expressed in piliated N. gonorrhoeae MS11 strains 27 . pilC expression is also subject to RecA-independent phase variation (on/off switching) due to frequent frameshift mutations occurring within homo-guanine tracts located within its signal peptide region 27 . PilC participates in pilus biogenesis as well as in host cell adherence, as pilC mutants prevent the formation of pili by negatively affecting their assembly process, which leads to the bacteria being unable to adhere to human epithelial cells 32 .
In addition to promoting attachment to host cells, type IV pili are also involved in bacterial twitching motility, biofilm formation, and DNA transformation 33 . N. gonorrhoeae is naturally competent for transformation in that it can take up exogenously produced Neisseria -specific DNA containing a 10-bp uptake sequence (GCCGTCTGAA; DUS) 34 . pilE mutations resulting in loss of pilus expression lead to transformation incompetence 28 , 35 . The binding and uptake of exogenous DNAs by N. gonorrhoeae requires type-IV-pili-structurally-related components, including ComP protein 36 , 37 . Despite sharing sequence similarity to PilE in the N-terminal domain, ComP was shown to be dispensable to Tfp biogenesis 36 . Instead the bacteria were unable to take up extraneous DNA; subsequent overexpression of ComP increased sequence-specific DNA binding, suggesting that ComP functions in the DNA binding step of transformation 37 . Recently, ComP has been shown to preferentially bind to DUS-containing DNAs via an electropositive stripe on its surface 38 with uptake of the DNA being facilitated by de-polymerization of the pilus structure through PilT hydrolytic activity 39 . The coordinated physical retraction and elongation of pili can lead to "twitching", a form of motility that propels the cell along a surface. Retraction is facilitated by PilT activity (an ATPase), whereas PilF protein promotes pilus elongation at the inner membrane 39 , 40 .
Por protein
The outer membrane porin protein, Por, is the most abundant protein in the gonococcus accounting for approximately 60% of the total protein content 1 . The molecular size of Por varies between strains, yet, within individual strains, it exists as only a single protein species 41 . Por has been used as the basis for serological classification of gonococci 41 with nine distinct serovars being identified 42 . Overall, there are two distinct structural classes (PorA and PorB) 42 , with the PorA subgroup tending to be associated with the more complicated aspects of the disease, whereas the PorB subgroup is more likely to be involved with uncomplicated mucosal infections 43 .
Porins allow the transport of ions and nutrients across the outer membrane and can also contribute to the survival of the bacteria in host cells 44 . Moreover, gonococcal Por protein has been shown to translocate from the outer membrane into artificial black lipid membranes 45 as well as into epithelial cell membranes, following attachment of the bacteria 46 . Por can also transfer into mitochondria of infected cells which leads to the formation of porin channels in the mitochondrial inner membrane, causing increased permeability 47 . This causes the release of cytochrome c and other proteins, leading to apoptosis of infected cells 48 . However, Por-induced apoptosis remains controversial. In direct contrast to events with the gonococcus, Neisseria meningitidis Por, which also interacts with mitochondria, apparently protects cells from undergoing apoptosis 49 . Interestingly, mitochondrial porins and Neisseria PorB share similar properties, with both protein species being capable of binding nucleotides and exhibiting voltage-dependent gating 50 . Por protein also modulates phagosome maturation by changing the phagosomal protein composition through the increase of early endocytic markers and the decrease of late endocytic markers, which ultimately delays phagosome maturation 51 .
Opacity-associated protein (Opa)
Opa proteins are integral outer membrane proteins and cause colonies to appear opaque due to inter-gonococcal aggregation when viewed by phase-contrast microscopy 52 , 53 , 54 . Opa proteins belong to a multigene family with a single gonococcal cell possessing up to 12 opa genes that are constitutively transcribed 55 , 56 . Each gene contains conserved, semivariable and 2 hypervariable regions, with the hypervariable segments of the proteins being located on the outside of the outer membrane 55 . Opa protein expression can undergo phase variation due to changing the numbers of pentameric repeat units (-CTCTT-) that are located within the leader peptide encoding region, which results in on/off switching of expression 57 . A single cell is capable of expressing either none to several different Opa proteins 57 , 58 .
Unlike pili, Opa expression is not required for the initial attachment of gonococci to the host. However, as an infection proceeds, Opa expression varies 58 , and Opa-expressing bacteria can be observed in epithelial cells and neutrophils upon re-isolation from infected human volunteers 59 , 60 . The invasive capacity of N. gonorrhoeae is determined by the differential expression of Opa 61 . Individual Opa proteins bind to a variety of receptors on human cells through their exposed hypervariable regions. The binding specificity for human receptors falls into two groups: OpaHS which recognize heparin sulfate proteoglycans 62 , 63 ; and, OpaCEA which recognize the carcinoembryonic antigen cell adhesion molecule (CEACAM) family that is comprised of the various CD66 molecules 64 , 65 , 66 , 67 . CEACAMs are the major receptors of Opa proteins and are expressed on many different cell types including epithelial, neutrophil, lymphocyte and endothelial cells 68 .
Lipooligosaccharide (LOS)
As with all Gram-negative bacteria, gonococci possess lipopolysaccharide in the outer membrane. Gonococcal LPS is composed of lipid A and core polysaccharide yet lacks the repeating O-antigens 1 . Accordingly, gonococcal LPS has been designated as lipooligosaccharide (LOS). Due to its surface exposure, gonococcal LOS is a primary immune target along with the major outer membrane protein Por 69 , 70 , 71 . Gonococcal LOS is also toxic to fallopian tube mucosa causing the sloughing off of the ciliatory cells 72 . The LOS oligosaccharide composition is highly variable both in length and in carbohydrate content. Consequently, heterogeneous LOS molecules can be produced by a single cell. However, distinct forms of LOS may be a prerequisite for infection in men 73 . The most common carbohydrates associated with isolated LOS molecules are lacto-N-neotetraose (Galβ(1-4)GlcNAcβ(1-3)Galβ(1-4)Glc) and digalactoside Galα(1-4)Gal and switching from one form to another occurs at high frequency 74 through phase variation of glycosyl transferases 75 , 76 . The variable oligosaccharide portions of LOS can also mimic host glycosphingolipids, thus promoting bacterial entry 74 . In addition, gonococcal LOS can also be sialylated which renders the bacteria resistant to serum killing 77 , 78 , 79 , 80 . Consequently, gonococcal LOS contributes to gonococcal pathogenicity by facilitating bacterial translocation across the mucosal barrier as well as by providing resistance against normal human serum 81 , 82 .
IgA protease
Immunoglobin A (IgA) protease is another virulence factor in N. gonorrhoeae 83 . Upon release from the cell, the protein undergoes several endo-proteolytic cleavages, leading to maturation of the IgA protease 84 . During an infection, the mature protease specifically targets and cleaves IgA1 within the proline-rich hinge region of the IgA1 heavy chain. The human IgA2 subclass is not cleaved by gonococcal IgA protease since it lacks a susceptible duplicated octameric amino acid sequence 85 . Neisseria IgA protease also cleaves LAMP1 (a major lysosome associated membrane protein), which leads to lysosome modification and subsequent bacterial survival 86 . Furthermore, iga mutants are defective in transcytosis of bacteria across an epithelial monolayer 87 .
PATHOGENESIS
Neisseria gonorrhoeae primarily colonizes the urogenital tract after sexual contact with an infected individual 88 . The gonococcus can exist as both an extracellular and intracellular organism, with the bulk of its genes being devoted to colonization and survival, due to the fact that it cannot survive outside of a human host 89 . Transmission is generally a consequence of sexual intercourse. Upon arrival into a new host, micro-colony formation commences on non-ciliated columnar epithelial cells approximately 1 to 2 hours post-infection 90 , 91 . Once the micro-colonies achieve a cell density of approximately 100+ diplococci, cytoskeletal rearrangement and host protein aggregation occurs, which leads to pilus-mediated attachment of the gonococcus to the CD46 host cell-surface receptor (Fig. 1) 89 , 92 . Once bound, the pilus structures on some organisms are retracted through PilE depolymerization 39 which promotes tighter contact with the host cells through Opa binding to the CEACAM receptors (Fig. 1) 65 , 66 . Upon CEACAM binding, actin polymerization and rearrangement is induced within the host cell which results in bacterial engulfment, transcellular transcytosis and release of the bacteria into the subepithelial layer (Fig. 1) 68 , 93 .
Figure 1. FIGURE 1: Schematic representation of a Neisseria gonorrhoeae infection.
1) Piliated, Opa-expressing gonococci interact with the mucosal epithelium. The thin, hair-like pilus appendages provide the initial contact with receptors on the surface of the mucosal cells.
2) Pili are then retracted which allows for more intimate, Opa-mediated attachment of the bacteria with the CD66 antigens located on the mucosal cells.
3) Following Opa-mediated attachment, the bacteria are engulfed and are internalized into the mucosal cells.
4) Following internalization, some bacteria can transcytose to the basolateral side of the mucosal epithelium.
5) Depending upon which Opa protein is being expressed, gonococci can also reside and survive inside of neutrophils.
6) Following transcytosis, gonococci can enter the bloodstream where heavy sialylation of lipooligosaccharide renders the bacteria serum resistant. This figure is based on 98 .
In vivo , the coordinated expression of pili and Opa varies considerably 94 . Organisms isolated from the male urethra generally co-express pili and one of several Opa proteins 58 . However, in women, Opa expression varies depending upon the stage of the menstrual cycle and whether or not the patient is taking oral contraceptives 94 . At mid-cycle, bacteria isolated from the cervix express Opa, whereas those isolated during menses tend to be Opa negative 17 . Moreover, organisms isolated from infected fallopian tubes are almost universally Opa negative, even though Opa expressing organism can be isolated from the cervix of the same patient 17 . These observations can perhaps be explained by the fact that cervical secretions during menstruation contain more proteolytic enzymes than during the follicular phase. Consequently, non-Opa expressing cells may be selected due to the extreme sensitivity of Opa proteins to trypsin-like enzymes. However, with the recent studies demonstrating Opa interactions with CECAM receptors, it has been observed that fallopian epithelial tube cell cultures do not appear to express CECAM receptors 95 . Nonetheless, in the absence of these receptors, gonococci were found to still adhere and invade. Consequently, CECAM expression, or the lack of it, possibly allows for in vivo phenotypic selection of distinct gonococcal populations on various tissues 96 . Overall, Opa expression does appear to increase gonococcal fitness within the female genital tract 97 . Generally, Opa expression is absent in most re-isolates from female disseminated infections.
Inflammation
The hallmark symptom of a non-complicated gonorrhea infection is a massive recruitment of neutrophils to the site of infection leading to the formation of a pustular discharge. Initially, Opa protein expression was suspected to be intimately involved in PMN stimulation 20 , 99 , 100 , 101 . Subsequently, it was shown that following attachment of gonococci to the mucosa, the pro-inflammatory cytokines IL-6 and TNF-alpha as well as the chemokine IL-8 are released leading to the recruitment of neutrophils 102 . In addition, upon arrival at the sub-epithelial layer, gonococci release LOS and lipoproteins which further stimulate cytokine production 103 as these outer membrane components are detected by Toll-like receptors (TLRs) on immune cells 104 . Host cells also respond to bacterial peptidoglycan fragments within outer membrane vesicles via cytoplasmic NOD-like receptors (NLRs) which also contribute to the secretion of additional pro-inflammatory cytokines 105 .
Despite the active recruitment of PMNs to a site of infection, gonococci can survive the oxidative and non-oxidative defense mechanisms (Fig. 1) 106 . Survival appears to correlate with gonococci selectively triggering Th17-dependent host defense mechanisms by modulating expression of IL-17 107 . Gonococci also must combat considerable oxidative stress by elaborating a number of different enzymes during the inflammatory response in order to detoxify superoxide anions (O2•−), hydrogen peroxide (H 2 O 2 ), and hydroxyl radicals (HO•) 108 , 109 . Gonococci must remove H 2 O 2 because in the presence of ferrous ions the Fenton reaction is initiated (Fe 2+ + H 2 O 2 → Fe 3+ + OH . + OH − ) which yields additional hydroxyl radicals 110 , 111 . Catalase is used by the gonococcus to eliminate H 2 O 2 (which significantly increases the organism’s ability to resist in vitro neutrophil killing) 112 in conjunction with a periplasmic cytochrome c peroxidase (Ccp) 110 . Normally, superoxide ions are removed by superoxide dismutase enzymes (SOD) which convert superoxide to H 2 O 2 and water. However, the majority of N. gonorrhoeae strains have no measurable SOD activity 108 , 109 , 110 , 111 , suggesting that oxidants may be removed via an alternative mechanism. It appears that N. gonorrhoeae utilize manganese ions (Mn 2+ ) to combat reactive oxygen species accumulation. Manganese accumulates within the cell through the Mn uptake system (MntABC), with Mn(II) and Mn(III) both scavenging superoxide and hydrogen peroxide molecules non-enzymatically. Furthermore, Mn(II)-pyrophosphate and Mn(III)-polyphosphate complexes are also effective in eliminating hydroxyl radicals that are formed via the Fenton reaction 110 .
The need for iron
Despite the problems associated with the Fenton reaction, iron is a vital nutrient, with pathogens expending considerable resources on scavenging the element from their human host. This becomes even more complicated during an infection, as the host responds to inflammation by limiting iron availability, as well as by decreasing free iron within the bloodstream 113 . Even though humans keep their iron sequestered in iron-protein complexes such as transferrin, lactoferrin, haemoglobin, and ferritin, the Neisseria are capable of scavenging iron from both transferrin and haemoglobin 114 , and express receptors for both transferrin and lactoferrin that provide a selective advantage within the host 115 . Because Neisseria do not produce siderophores, they must directly extract iron from transferrin. To achieve this, the iron transport system consists of two large surface proteins, transferrin binding protein A (TbpA) and transferrin binding protein B (TbpB), with both of these proteins being found in all clinical isolates of pathogenic Neisseria 116 . TbpA is an outer membrane transporter essential for iron uptake that binds both apo- and iron-containing transferrin with similar affinities, whereas TbpB, a surface-exposed lipoprotein, only associates with iron-bound transferrin 117 . As the affinity of the bacterial receptor for iron is similar to transferrin’s affinity, this enables the gonococcus to compete with the host for this necessary nutrient 118 . Subsequently, it was shown that the expression of the transferrin receptor was absolutely required for gonococcal infectivity 119 .
Serum resistance
Bactericidal antibody-mediated killing was found to vary greatly between patients presenting genital infections 120 . Subsequently, it was soon recognized that gonococcal surface components were the primary targets of antibody-dependent complement killing, with LPS-specific antibodies being the most effective at inducing bactericidal responses 121 . Two forms of serum resistance were initially described; stable and unstable serum resistance 77 , 122 . Unstable serum resistance is due to the modification of gonococcal LOS through the addition of sialic acid molecules to terminal galactose residues using cytidine 5’-monophosphate N-acetylneuraminic acid (CMP-NANA) which is abundant in human serum, as well as in various mucosal secretions and within professional phagocytes. Sialic acid transfer uses the conserved outer membrane-located enzyme 2,3-sialyltransferase 79 . Sialylation of LOS mediates both the entry of gonococci into host mucosal cells as well as influencing bacterial resistance to killing by complement 82 . Gonococcal cells harboring lightly sialylated LOS molecules are able to invade host epithelial cells more efficiently than heavily sialylated-LOS variants. However, lightly sialylated-LOS expressing cells are more susceptible to complement-mediated killing, whereas, heavy sialylation of LOS renders the bacteria resistant to normal human serum by masking the target sites for bactericidal antibodies 78 , 80 which prevents the functional activation of the complement cascade (Fig. 1) 81 .
In contrast, stable serum resistance appears to be caused through the faulty insertion of the C5b-C9 membrane attack complex in serum resistant strains 123 , 124 , 125 . Accompanying this defect in deposition, blocking antibody is also thought to cause the C3 complement component to be loaded onto a different site on the outer membrane such that it again hinders bactericidal killing 126 . Clearly, complement resistance is important for organisms causing a disseminated infection, but its value is less clear for those organisms causing a mucosal infection. However, seminal plasma does contain an inhibitor of complement activation suggesting that there is some complement activity at the mucosa 127 .
As indicated previously, the major outer membrane protein, Por, exists in two forms, Por1A and Por1B, with Por1A-expressing gonococci being most often associated with disseminated infections 42 , 43 . Por1A-expressing gonococci also bind complement factor H more efficiently, and, as factor H down-regulates alternative complement activation, such binding helps explain serum resistance in these disseminated strains 128 . Furthermore, it also helps explain species-specific complement evasion 129 . Por protein also influences activation of the classical complement pathway, as Por binds to the C4b-binding protein, which again down-regulates complement activation 130 . Consequently, as factor H and C4b-binding sites on the Por proteins impede functional complement deposition these may need to be modified in vaccine preparations as this may help alleviate problems associated with serum resistance 131 .
Active immunity
It has long been known that gonorrhea does not elicit a protective immune response and nor does it impart immune memory. Consequently, individuals can become repeatedly infected. Nonetheless, specific antibodies are generated within the genital tract that inhibit adherence to the mucosal epithelium, yet their persistence appears to be short-lived 18 , 132 . Overall, the immune response to an uncomplicated genital infection remains modest 133 .
The general unresponsiveness to an infection appears to stem from the organism being able to manipulate the host cell response. Transient decreases in T-cell populations occur within the bloodstream and appear to reflect Opa protein interactions with CD4 + T-cells which suppresses T-cell activation 134 . Moreover, in contrast to Opa-mediated interactions with CEACAM antigens on other cell types, Opa-CEACAM1 T-cell interactions do not appear to cause the internalization of bacteria into the T-cells. This then leads to a dynamic re-cycling response with the T-cells that ultimately suppresses an immune response 135 . Likewise, Opa-CEACAM1 interactions on B lymphocytes also inhibit antibody production 133 , 136 . Even with dendritic cells, Opa-CEACAM1 interactions do not stimulate internalization 136 . Instead, engulfment by dendritic cells is mediated through LOS interaction with DC-SIGN antigens. Consequently, as LOS molecules vary in composition, this allows the gonococcus a further opportunity for immune evasion 137 . LOS molecules often activate immune cells through interaction with Toll-like receptors. However, LOS deacylation can moderate an immune response following interaction with its cognate Toll-like receptor leading to B-cell proliferation where antibody production is down-regulated 138 .
Recently, an artificial estradiol-induced mouse infection model has been developed for gonococcal infections that allows for in vivo assessment 139 . However, major differences exist between the human and mouse female genital tract. For example, the pH of the mouse vagina is higher, there is no comparable menstral cycle, fewer anaerobic commensal bacteria are present, and as the mice need to be treated with antibiotics, this aspect dramatically changes the resident flora 140 . Nonetheless, the mouse infection model has yielded several interesting observations. Using the model, gonococci have been shown to moderate the murine innate immune response by stimulating IL-17 release from TH17 cells which subsequently effects other cells 107 . In conjunction with transforming growth factor beta (TGF-beta), this coupled cytokine presence suppresses Th1/Th2 adaptive responses 141 . Therefore, as the genital tract is rich in TGF-beta, gonococci naturally inhabit an immunosuppressive environment 142 . Again, LOS and Opa expression play a major role in these responses, as LOS drives the Th17 response with Opa negatively impacting the Th1/Th2 responses 142 . Further manipulation of the host response is also seen with gonococcal activation of IgM-specific memory B-cells in a T-independent manner. Consequently, this elicits a non-specific polyclonal immunoglobulin response without generating specific immunologic memory to the gonorrhea infection 143 . Recently, human CEACAM transgenic mouse models have been developed for studying gonococcal in vivo infections 144 , 145 . With these more refined models, gonococci were shown to readily infect and cause inflammation in the transgenic animals and that Opa-CEACAM interactions dramatically reduced exfoliation of the murine mucosal surface. As gonococci bind to human CR3 (hCR3) integrin to invade cervical cells and that human factor H bridges the interaction between the bacteria and hCR3, then future transgenic mouse models, expressing both hCR3 and human factor H, may further mimic a bona fide gonococcal infection in vivo .
Antigenic variation
Neisseria gonorrhoeae can survive either as an extracellular organism, or, alternatively, as an intracellular organism within a variety of different cell types. Which state the organism enters depends largely on which surface components are expressed and whether these components are chemically modified or not. N. gonorrhoeae can modulate expression, or, the chemical character of its surface components either by phase variation, or, by antigenic variation 25 . Generally, phase variation is a consequence of frame-shifting within a gene which leads to random switching between on/off states, whereas antigenic variation leads to changes in the chemical composition of some structural component. Therefore, each gonococcal cell can differentially express distinct surface antigens, in various chemical forms, which hinders recognition by host antibodies, facilitates multiple lifestyles 25 and helps explain the lack of efficacious vaccines to protect against a gonorrheal infection 21 .
From genome analysis, 72 putative genes were identified that have the capacity to undergo phase variation 146 . Consequently, the stochastic expression of various surface components leads to the emergence of micro-populations that allows colonization within unique environmental niches 147 . Pilus expression can undergo on/off switching due to frameshifting either within the pilE gene 35 , or, within the pilC gene 27 . Similarly, LOS variation depends upon frameshifting within various glycosyl transferase genes which leads to the random acquisition of various sugar moieties on a varying LOS molecule 75 , 76 . Opa expression relies exclusively on phase variation, as a series of pentameric repeats (-CTCTT-) reside towards the 5' end of each opa gene 57 . Consequently, the addition or subtraction of a repeat(s) will bring each individual opa gene either in or out of frame. As expression of individual Opa proteins influence the cellular tropism of the organism with regards to internalization into either mucosal or lymphocytic cells, opa phase variation allows variable gonococcal populations to be established that have the potential to internalize into whatever cell becomes available 56 , 61 . Consequently, phase variation confers a degree of fitness on the organism for a specific environment, yet provides little with respect to bona fide immune evasion.
Antigenic variation on the other hand confers remarkable immune evasion. Antigenic variation occurs extensively within the pil system as well as in some other minor systems ( maf and fha ) 26 . Gonococci possess multiple variable pil genes; some are deemed silent ( pilS ) and serve as storage loci for variable pil sequence, and act in conjunction with a single expression locus, pilE , which encodes the PilE polypeptide. Recombination frequently occurs between pilE and an individual pilS leading to changes in the chemical composition of PilE. It is estimated that PilE can assume 108 chemical forms 148 which helps thwart an efficacious immune response due to its prominent surface location. Therefore, despite the fact that anti-pilus antibodies can be detected within the genital tract such antibodies do not recognize heterologous strains thus allowing for reinfection of an individual 18 .
It is in the coordinated variation of these various surface components that allow gonococci to develop adaptive strategies where the organism can exist either externally or internally during an infection (Fig. 1). When gonococci reside externally, the organisms are generally piliated, with PilE undergoing antigenic variation which negates the various antibody clearing strategies. When coupled with the appropriate LOS composition, these organisms can also become heavily sialylated, which impedes serum killing, thus facilitating extra-cellular growth. In contrast, internalization into host cells requires the retraction of pili causing the cells to become non-piliated. When coupled with phase variation of Opa expression and a non-sialylatable LOS phenotype, the gonococcus can translocate across the mucosal epithelium at an initial stage of the infection and ultimately reside internally within various cell types 25 . Eventually, infected host cells will undergo apoptosis, releasing bacteria back onto the mucosal lining, where in the presence of seminal plasma the appropriate cell surface reappears to facilitate transit into a new host 149 .
Vaccine development
Vaccine development for sexually transmitted diseases has long been a goal of the scientific community 150 , 151 . However, given the extensive antigenic variation displayed by N. gonorrhoeae , coupled with suppression and manipulation of the host immune response, progress has been severely impeded. Nonetheless, in the mouse infection model, if Th1 responses can be induced, an infection will clear and immune memory can be established 152 . Consequently, incorporating Th1-inducing adjuvants within any vaccine preparation may be crucial for success in this endeavor.
Two outer membrane proteins have come under considerable scrutiny as potential vaccine components; pilus constituents and the major outer membrane protein, Por. Because anti-pili antibodies were detected in vaginal secretions following an infection 18 , this led to the early development of a parenteral pilus vaccine. Unfortunately, administration of this vaccine afforded partial protection only to homologous strains. Moreover, it also showed poor immunogenicity and did not stimulate an adequate antibody response at the site of infection 21 , 153 , 154 . Consequently, other antigens were explored as potential vaccine candidates. As neisserial Por proteins can serve as adjuvants to B-cells, as well as stimulate Por-specific circulating Th2-cells that appear to migrate to mucosal surfaces, Por has come under considerable scrutiny 155 , 156 . Por is also capable of stimulating dendritic cells where activation depends on Toll-like receptor 2. Therefore, as Por composition is relatively stable, this protein has become a promising vaccine candidate, especially if Th1-inducing adjuvants and Toll-like 2-inducing adjuvants can be included within any "designer" vaccine preparation 157 , 158 , 159 .
However, a problem exists in the development of any vaccine in that antibodies within normal human serum bind to the gonococcal outer membrane protein Rmp with binding apparently, having important consequences with regard to serum resistance for the organism 160 , 161 . The presence of cross-reactive Rmp antibodies also facilitates transmission 161 and women with Rmp antibody titers appear at an increased risk for infection 162 . As the Rmp protein is in close association with Por protein 163 it would appear to be imperative that Rmp protein is excluded from any Por-based vaccine preparation. Nonetheless, a quiet optimism now pervades the field that an anti-gonococcal vaccine may be around the corner 152 .
MOLECULAR EPIDEMIOLOGY - A HISTORICAL REVIEW
Auxotyping and serotyping - 70’s through the early 80’s.
As public health decisions regarding transmissible pathogenic diseases rely heavily on epidemiological surveillance, it became necessary to accurately identify and characterize the different circulating strains of N. gonorrhoeae 164 . Initially, isolates were typed through growth responses on chemically defined media 165 , 166 or by serotyping using common protein antigens or lipopolysaccharide 41 , 42 , 43 , 167 . Consequently, the identification of different auxotypes allowed different N. gonorrhoeae strains to be typed with respect to disease severity 168 , 169 . Subsequent Por-based serotyping allowed isolates to initially be grouped into two structurally related forms 41 , 44 , 170 , which was then further refined using enzyme-linked immunosorbant assays to eventually define nine different Por-based serotypes 171 .
Attempts were then made to differentiate isolates that caused uncomplicated, localized infections and those that caused disseminated gonococcal infections (DGI) 172 . DGI phenotypes included an increased sensitivity to penicillin 173 , unique nutritional requirements 168 coupled with serum resistance which led to increased virulence of DGI isolates 172 . Subsequently, it has been shown that the majority of DGI isolates belonged to two distinct serotypes 43 , 174 .
The emergence of antibiotic-resistant strains of N. gonorrhoeae identified a need to determine modes of antibiotic resistance among strains in order to monitor the development of new resistance genes, the lateral transfer of resistance genes, or the spread of resistance strains among the population. Early genetic mapping identified several genes involved in antibiotic resistance 175 . Through epidemiologic studies and characterization of penicillinase-producing N. gonorrhoeae (PPNG), it was determined that two indeendent strains of PPNG arose in geographically separate populations; both carried the resistance gene on distinct plasmids, with one strain (linked to the Far East) being more prevalent than the strain linked to West Africa 176 . Analysis of PPNG strains demonstrated that their introduction into the United States was due to returning military personnel from the Far East. Travel also contributed to global spread of these strains, as patients would encounter penicillin-resistant β-lactamase-producing N. gonorrhoeae following rendezvous with overseas prostitutes, which would in turn often transmit them to local prostitutes, thereby continuing their spread 169 , 177 .
Such analysis of clinical isolates indicated that distinct reservoirs of infection could be detected based upon sexual preference. Studies revealed that homosexual men had a lower incidence of asymptomatic urethral infections and DGIs, yet more frequently acquired infections by strains that were more resistant to penicillin G, which at the time, accounted for the high failure rate of this antibiotic for rectal infections 178 . Also, reservoirs for certain PPNG outbreaks could be traced back to female prostitutes, as these strains were largely absent from the homosexual community. Further epidemiological studies were able to identify gonococci that were exclusively present in both heterosexual men and women, or within homosexual male communities, thus defining sources of infection between male and female partners, prostitution and/or same sex partners 169 .
"Core group" hypothesis - late 70’s through the 80’s
As previous gonorrheal infections provide little to no immunity to subsequent infections, an alternative model for gonorrhea transmission was proposed, t suggesting that all cases of the disease are caused by a core group of individuals 179 . This "core group" hypothesis, was later reinforced by the emergence and spread of PPNG from the Far East 169 , 179 and through clinical investigations in the United States 180 , 181 . The persistence of isolates within a community was proposed to be due to a number of factors including the tendency for these strains to cause asymptomatic infections, or, alternatively, to have long incubation times prior to the onset of symptoms, which provided support to the theory that a core group of transmitters, most likely prostitutes, transmit the disease to many sexual partners 169 . Epidemiological studies revealed that a substantial group of individuals (33%) admitted to continual sexual engagement even with the knowledge of potential exposure, or, worse, even after the onset of symptoms, and that men with new or multiple sex partners were more likely to contract gonorrhea 182 , 183 . Consequently, five sociological trends were identified that assisted the rise of gonorrhea infections: 1) frequent changes in sex partners, 2) increased population mobility, 3) increasing gonococcal resistance to antibiotics, 4) decreased condom, diaphragm and spermicide use, and 5) increasing the use of oral contraceptives 184 .
Linkage disequilibrium - 1993
With the widespread use of serological typing, coupled with the desire for vaccine development, the classification and characterization of gonococcal strains invariably focused on investigating surface exposed antigens 185 . However, the combination of auxotyping and serotyping proved to be unreliable, as these techniques did not always provide adequate resolution 186 . As most pathogens are clonal with a disposition towards linkage disequilibrium, this property generally allows for classification based upon nucleotides that are present at variable sites, which in turn allows the serology, pathogenicity, host specificity and the presence of virulence genes to be mapped 185 , 187 . However, panmictic microorganisms, such as the gonococcus, that undergo mutation and frequent recombinational exchanges, do not allow stable clones to emerge due to the randomization of alleles within a population. Consequently, this complicates epidemiological characterization. Also, as surface-exposed antigens that are used for serotyping also tend to evolve rapidly due to strong diversifying selection placed on them by the host immune system, this further compounds the problem 185 . Given the above problems, it became necessary to index genes that only undergo neutral variation in order to investigate population structure, which led to analysis being focused on housekeeping genes involved in central metabolism 188 . Consequently, novel methods of molecular typing were then devised to define outbreaks based on either local or global epidemiology 164 .
Multilocus enzyme electrophoresis (MLEE) - 90’s
The advent of multilocus enzyme electrophoresis (MLEE) allowed for the presence or absence of linkage disequilibrium within a population to be monitored via deviations between multiple chromosomal alleles 188 , 189 , 190 , 191 . Indeed, this approach allowed for global epidemiological studies and permitted the identification of strains with an increased tendency to cause disease 164 . Statistical analysis performed on the electrophoretic types of 227 global N. gonorrhoeae isolates provided evidence of a panmictic population structure, as no single pair of alleles was statistically significant for linkage disequilibrium. Additionally, it was determined that the genetic variability of isolates obtained from the same geographic location was as great as that found between all geographic locations that were analyzed. Consequently, it was concluded that the propensity for individual hosts to carry more than one genotype of N. gonorrhoeae , combined with natural competence for DNA transformation, promoted the highly panmictic nature of this pathogen 189 .
Multilocus sequence typing (MLST) - 90’s
However, MLEE had limitations as it could only detect a small proportion of mutations through differences in electrophoretic mobility 164 , 185 . Therefore, nucleotide sequencing of the core gene set was then introduced leading to multilocus sequence typing (MLST) 164 . This proved to be extremely effective at detecting relationships between identical or closely related isolates by characterizing them on the basis of sequence variation 164 , 192 . While MLST typing could be readily applied to N. meningitidis isolates, it was initially thought that clinical isolates of N. gonorrhoeae could not be used, as gonococcal housekeeping genes appeared to be homologous 164 , 185 , 193 . Also, as frequent recombination occurred within the organism, it was initially believed that the genetic relatedness of distant isolates may become obscured 194 . However, recombinant exchanges must accrue over long time periods for relationships to be masked, and as the field of molecular epidemiology is only concerned with very short evolutionary time scales, any correlations drawn are unlikely to be skewed by recombination 192 . Therefore, MLST studies did show that N. gonorrhoeae isolates could be typed using the same methods applied to N. meningitidis 164 and N. lactamica 18 , 195 . It was through comparison of MLST data among the Neisseriae , that it was postulated that minimal interspecies recombination actually occurs among the housekeeping genes 186 .
eBURST - 2000’s
Typically, MLST allelic profiles were placed into a matrix of pairwise differences which allows for detection of identical or closely related isolates. However, these do not provide the necessary information on the evolutionary descent of genotypic clusters, nor do they identify the founder genotype 192 . Additionally, in bacterial species such as N. gonorrhoeae that undergo frequent recombination, any relatedness that may be implied through the use of pairwise differences is highly suspect and most likely may not be phylogenetically relevant 196 . To account for these concerns, the BURST (based upon related sequence types) algorithm was designed to analyze microbial MLST data by assigning defined sequence types (STs) to lineages which allowed the identification of a putative founder genotype 197 .
The program was further refined with the development of the eBURST algorithm, which differentiates large MLST datasets based on isolates with the most parsimonious descent pattern from the probable founder, and allows for the identification of clone diversification yet also provides insight into the emergence of clinically relevant isolates 192 . Initially, eBURST was used for analysis of quinolone-resistant N. gonorrhoeae (QRNG) 198 . Previous epidemiological studies of quinolone resistance strains of N. gonorrhoeae could not determine if distinct isolates arose due to variation of an original strain or if multiple strains were concomitantly introduced into a specific geographic location 198 , 199 , 200 , 201 . eBURST analysis determined the total number of QRNG strains that entered a country, the divergence of loci, and the time period during which the founder strains evolved 198 . With the combination of MLST and eBURST analysis, disease isolates could now be defined with regard to distribution, population structure, and evolution 202 . Consequently, the origins of pathogenic strains could now be determined as well as how bacterial populations respond to antibiotics and vaccines through analysis of recent evolutionary changes 203 .
CHEMOTHERAPY
Neisseria gonorrhoeae is rapidly evolving and has developed resistance to all previous and current antimicrobials. The recent emergence of multidrug resistant gonococcal isolates in Japan 204 , France 205 , and Spain 206 has provoked major concerns in public health circles worldwide, especially as drug resistance is spreading rapidly 207 . Consequently, we may be entering an era of untreatable gonorrhea. Medications such as penicillin, and later, the fluoroquinolines, have each been used to treat gonorrhea in the past, however, resistance to these antimicrobial agents quickly developed, leaving limited options for gonococcal treatment 208 . Currently, third generation extended-spectrum cephalosporins (ESCs); which include ceftriaxone (injectable form) and cefixime (oral form) are being prescribed. However, resistance to ESCs has also emerged with resistant isolates having been reported in 17 different countries 209 , 210 .
The recent emergence of the first N. gonorrhoeae "superbug" strain in Japan (H041, which was later assigned to MLST ST7363) has been shown to exhibit extremely high-level resistance to all ESCs, including cefixime (MIC= 8 µg/ml), and ceftriaxone (MIC= 2-4 µg/ml) as well as to almost all other available therapeutic antimicrobials 204 . Since the isolation of the H041 strain, other extensive drug resistance (XDR) strains have also been isolated in Quimper, France (F89 strain) 205 as well as in Catalonia, Spain 206 , and both share considerable genetic and phenotypic similarity to the Japanese H041 strain. Unfortunately, transmission of these strains is augmented by the fact that XDR strains have been isolated from commercial sex workers, homosexual men, sex tourists, long distance truck drivers, and people undergoing forced migration, suggesting that these strains have the potential to spread globally 207 .
N. gonorrhoeae are exceptional bacteria that can rapidly evolve to promote adaptation and survival within different niches of the human host. This is facilitated by their natural competence which allows DNA uptake from the environment via transformation, as well as by engaging in bacterial conjugation. Consequently, gonococci can acquire various different types of antimicrobial resistance (AMR), which include drug inactivation, modification of drug targets, changing bacterial permeability barriers, and increasing efflux properties 208 , 209 . The acquisition of AMR genes was initially thought to occur within commensal Neisseria spp. that reside in the pharynx, as pharyngeal organisms are often exposed to antimicrobials, with the fixed mutations then being transferred to gonococci that mingle with the commensal bacteria 211 . Neisseria can also obtain AMR through spontaneous mutations, although such events are comparatively rare. Many resistance determinants originate through the accumulation of chromosomal mutations, with only two known plasmid-borne genes having been described; penicillin resistance associated with the blaTEM plasmid 212 , 213 , 214 and tetracycline resistance associated with the tetM plasmid 215 . Penicillinase-producing strains of Neisseria gonorrhoeae were first isolated in Southeast Asia and in sub-Saharan Africa 176 . However, less than one percent of gonococcal clinical isolates in the US contain the β-lactamase gene, indicating that the major mechanism of penicillin resistance appears to result from accumulation of chromosomal mutations over time 214 . Interestingly, the N. gonorrhoeae tetM conjugative plasmid 216 is not only self-transmissible but is also responsible for transfer of the β-lactamase plasmids to other gonococci, other Neisseria spp., and E. coli 217 , 218 .
Chromosomal-mediated resistance to penicillin, as well as to other ESCs, generally involves modification of the penicillin binding proteins (PBP) coupled with mutations that enhance the efflux and decrease the influx of antimicrobials. Penicillin-resistant gonococcal strains typically contain 5 to 9 point mutations in the penA gene which encodes PBP2, the primary lethal target of the β-lactam antimicrobials 219 , 220 . Penicillin and ESC minimum inhibitory concentrations (MICs) can also be elevated in strains carrying mtrR and penB mutations which increase efflux and decrease influx of the antimicrobials, respectively 204 , 205 . Surprisingly, synergy between mtrR and penB mutations appears to have very little impact on resistance to cefixime which is mainly conferred by penA mosaic alleles 221 .
Once acquired, resistance determinants contributing to decreased susceptibility or resistance to certain antibiotics are stably carried within Neisseria populations even when the antibiotic is withdrawn from treatment regimens 208 . The persistence of resistance determinants also suggests that these factors do not cause a negative impact on the biological fitness of the gonococcus. In fact, antibiotic resistance can be linked with enhanced fitness as demonstrated with the MtrCDE efflux system that contributes to gonococcal virulence and survival during an infection 222 , 223 . This efflux pump can recognize and expel not only hydrophobic antibiotics such as penicillin, ESCs, macrolides, tetracycline, and ciprofloxacin 224 , 225 , 226 , but also antimicrobial compounds from the innate host response such as antimicrobial peptides, bile salts, and progesterone, allowing the bacteria to survive within host cells 227 .
Future directions
Due to the lack of an efficacious vaccine, control of gonococcal infections relies on appropriate antibiotic treatment, coupled with prevention, proper diagnosis, and epidemiological surveillance. Recently, novel dual antimicrobial therapy, e.g. ceftriaxone and azithromycin 228 , 29 or gentamicin and azithromycin 230 combination treatment, has been evaluated for treatment of uncomplicated gonorrhea. However, the emergence of concomitant resistance to the available antimicrobials has again compromised such an approach 207 , 208 , 228 , 231 .
Previously developed antibiotics, including gentamicin, solithromycin, and ertapenem, are also now being considered as clinical isolates show a high degree of sensitivity to these antibiotics in vitro 232 , 233 . The carbapenem, ertapenem, is potentially an option for ceftriaxone -resistant N. gonorrhoeae as these strains display relatively low MICs when treated with this agent 234 . However, regimens with ertapenem are only applicable if ertapenem and ceftriaxone do not share the same resistance mechanism such as strains carrying certain penA , mtrR , and penB mutations which coincided with increased carbapenem MICs 209 , 234 . Consequently, using these antimicrobials may only provide a short-term solution for combating multidrug-resistant gonorrhea 207 .
To counteract this problem, new antibiotics are being developed for anti-gonococcal therapy. The novel macrolide-family of antibiotics, such as bicyclolides modithromycin (EDP-420) and EDP-322, display high activity against azithromycin-, ESC-, and multidrug-resistant gonococcal isolates in vitro . However, these macrolide drugs appear to cause some cross-resistance with high-level azithromycin resistance 235 . The tetracycline derivatives, glycylcycline tigecycline and fluorocycline eravacycline (TP-434), have also been shown to be effective against ceftriaxone-resistant gonococci in vitro , yet, concerns remain regarding their usage and effectiveness 236 , 237 . Recently, new broad-spectrum fluoroquinolones, such as avarofloxacin (JNJ-Q2) 238 , delafloxacin, sitafloxacin 239 , and WQ-3810 240 , have displayed high potency against multidrug-resistant gonococcal isolates in vitro including ciprofloxacin-resistant strains. Finally, the lipoglycopeptide dalbavancin and 2-acyl carbapenems, SM-295291 and SM-369926, are among potential antimicrobials that can be used in gonorrhea treatment to a limited extent 241 .
Current research has centered on exploring novel antimicrobials or compounds designed to inhibit new targets. Among these newly developed agents are a protein inhibitor (pleuromutilin BC-3781), a boron-containing inhibitor (AN3365) 242 , efflux pump inhibitors, which enhance susceptibility to antimicrobials, host innate defense components and toxic metabolites 226 , 243 , non-cytotoxic nanomaterials 244 , host defense peptides- LL-37 (multifunctional cathelicidin peptide) 245 , molecules that mimic host defensins, LpxC inhibitors 246 , species-specific FabI inhibitors (MUT056399) 247 , and inhibitors of bacterial topoisomerases (VT12-008911 and AZD0914) both of which target alternative sites other than the fluoroquinolone-binding site 248 . Importantly, all these compounds possess potent in vitro activity against multidrug-resistant gonococcal strains 208 , 249 . The novel spiropyrimidinetrione antibacterial compound (AZD0914) which inhibits DNA biosynthesis 250 appears to be extremely promising, as no emerging resistance has been observed in diverse multidrug-resistant gonococcal isolates 235 . Consequently, AZD0914 is being seriously considered for its potential use as future oral treatment for gonococcal infections especially as it lacks cross-resistance exhibited by other fluoroquinolone antibiotics 251 .
Ideally, the future treatment for gonorrhea will rely on individually-tailored regimens as clinical isolates will hopefully be rapidly characterized by novel phenotypic AMR tests and rapid genetic point-of-care antimicrobial resistance tests. Unfortunately, no commercial molecular diagnostic kit is currently available to detect AMR determinants in gonococci, with the current genetic assays lacking sensitivity and specificity 249 , 252 . Meanwhile, healthcare initiatives need to be immediately undertaken to postpone the further widespread dissemination of ceftriaxone-resistant N. gonorrhoeae strains. These measures should include conducting AMR surveillance on global, national, as well as local scales, identifying treatment failures, monitoring the susceptibility of gonococcal isolates to prescribed antibiotics, and using appropriate and effective antibiotics with optimized quality and doses in gonorrhea treatment regimens 209 .
Funding Statement
Dedicated to the memory of John Swanson.
- 1. Britigan BE, Cohen MS, Sparling PF. Gonococcal infection: a model for molecular pathogenesis. N Eng J Med. 1985;312:1683–1694. doi: 10.1056/NEJM198506273122606. [ DOI ] [ PubMed ] [ Google Scholar ]
- 2. US Department of Health and Human Services Sexually Transmitted disease Surveillance. Centers for Disease Control and Prevention. 2013 [ Google Scholar ]
- 3. World Health Organization Department of Reproductive Health and Research Global incidence and prevalence of selected curable sexually transmitted infecions - 2008. 2012 Available: http://apps.who.int/iris/bitstream/10665/75181/1/9789241503839_eng.pdf?ua=1 . [ Google Scholar ]
- 4. Wasserheit JN. Effects of changes in human ecology and behavior on patterns of sexually transmitted diseases, including human immunodeficiency virus infection. Proc Natl Acad Sci U S A. 1994;91(7):2430–2435. doi: 10.1073/pnas.91.7.2430. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 5. Edwards JL, Butler EK. The pathobiology of Neisseria gonorrhoeae lower female genital tract infection. Front Microbiol. 2011;(2):102–115. doi: 10.3389/fmicb.2011.00102. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 6. Sutton MY, Sternbergy M, Zaidi A, St Louis ME, Markowitz LE. Trends in Pelvic Inflammatory Disease hospital discharges and ambulatory visits, United States, 1985-2001. Sex Transm Dis. 2005;32(12):778–784. doi: 10.1097/01.olq.0000175375.60973.cb. [ DOI ] [ PubMed ] [ Google Scholar ]
- 7. French CE, Hughes G, Nicholson A, Yung M, Ross JD, Williams T, Soldan K. Estimation of the rate of pelvic inflammatory disease diagnoses: trends in England, 2000-2008. Sex Transm Dis. 2011;38(3):158–162. doi: 10.1097/olq.0b013e3181f22f3e. [ DOI ] [ PubMed ] [ Google Scholar ]
- 8. Ross JDC, Hughes G. Why is the incidence of pelvic inflammatory disease falling? BMJ. 2014;348(feb135):g1538. doi: 10.1136/bmj.g1538. [ DOI ] [ PubMed ] [ Google Scholar ]
- 9. Nathan L, Decherney AH, Pernoll ML. Lange Medical Books. New York: McGraw-Hill; 2003. Current obstetric & gynecologic diagnosis & treatment. 9th edition . [ Google Scholar ]
- 10. Cohen MS, Hoffman RA, Royce RA, Kazembe P, Dyer JR, Daly CC, Zimba D, Vernazza PL, Maida M, Fiscus A, Eron JJJ. Reduction of concentration of HIV-1 in semen after treatment of urethritis: implications for prevention of sexual transmission of HIV-1. Lancet. 1997;349(9069):1868–1873. doi: 10.1016/S0140-6736(97)02190-9. [ DOI ] [ PubMed ] [ Google Scholar ]
- 11. Hastings CJ. Public Health Aspects of Venereal Diseases. Public Health J. 1917;8:37–41. [ Google Scholar ]
- 12. Kellogg DSJ, Deacon WL, Brown WE, Pirkel CI. Neisseria gonorrhoeae. I. Virulence genetically linked to clonal variation. . J Bacteriol. 1963;85(6):1274–1279. doi: 10.1128/jb.85.6.1274-1279.1963. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 13. Kellogg DSJ, Cohen IR, Norins LC, Schroeter AL, Reising G. Neisseria gonorrhoeae II. Colonial variation and pathogenicity during 35 months in vitro. . J Bacteriol. 1968;96(3):596–605. doi: 10.1128/jb.96.3.596-605.1968. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 14. Swanson J, Kraus SJ, Gotschlich EC. Studies on gonococcus infection. I. Pili and zones of adhesion: their relationship to gonococcal growth patterns. J Exp Med. 1971;134(4):886–906. doi: 10.1084/jem.134.4.886. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 15. Swanson J, Robbins K, Barrera O, Corwin D, Boslego J, Ciak J, Blake M, Koomey JM. Gonococcal pilin variants in experimental gonorrhea. J Exp Med. 1987;165(5):1344–1357. doi: 10.1084/jem.165.5.1344. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 16. Pearce WA, Buchanan TM. Attachment role of gonococcal pili: optimum conditions and quantification of adherence of isolated pili to human cells in vitro. J Clin Invest. 1978;61(4):931–943. doi: 10.1172/JCI109018. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 17. Draper DL, James JF, Brooks GF, Sweet RL. Comparison of virulence markers of peritoneal and fallopian tube isolates with endocervical Neisseria gonorrhoeae isolates from women with acute salpingitis. Infect Immun. 1980;27(3):882–888. doi: 10.1128/iai.27.3.882-888.1980. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 18. Tramont EC, Ciak J, Boslego J, McChesney DG, Brinton CC, Zollinger W. Antigenic specificity of antibody in vaginal secretions during infection with Neisseria gonorrhoeae. J Infect Dis. 1980;142(1):23–31. doi: 10.1093/infdis/142.1.23. [ DOI ] [ PubMed ] [ Google Scholar ]
- 19. Densen P, Mandell GL. Gonococcal interactions with polymorphonuclear neutrophils. J Clin Invest. 1978;62(6):1161–1171. doi: 10.1172/JCI109235. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 20. Virji M, Heckels JE. The effect of protein II and pili on the interaction of Neisseria gonorrhoeae with human polymorphonuclear leukocytes. J Gen Microbiol. 1986;132:503–512. doi: 10.1099/00221287-132-2-503. [ DOI ] [ PubMed ] [ Google Scholar ]
- 21. Boslego JW, Tramont EC, Chung RC, McChesney DG, Ciak J, Sadoff JC, Piziak MV, Brown JD, Brinton CC, Wood SW, Bryan JR. Efficacy trial of a parenteral gonococcal pilus vaccine in men. Vaccine. 1991;9(3):154–162. doi: 10.1016/0264-410X(91)90147-X. [ DOI ] [ PubMed ] [ Google Scholar ]
- 22. Freitag NE, Seifert HS, Koomey M. Characterzation of the pilF-pilD pilus-assembly locus of Neisseria gonorrrhoeae. Mol Microbiol. 1995;16(3):575–586. doi: 10.1111/j.1365-2958.1995.tb02420.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 23. Forest KT, J A Tainer. Type-4-pilus-structure: outside to inside and top to bottom- a minireview. Gene. 1997;192(1):165–169. doi: 10.1016/S0378-1119(97)00008-5. [ DOI ] [ PubMed ] [ Google Scholar ]
- 24. Hagblom P, Segal E, Billyard E, So M. Intragenic recombination leads to pilus antigenic variation in Neisseria gonorrhoeae. Nature. 1985;315:156–158. doi: 10.1038/315156a0. [ DOI ] [ PubMed ] [ Google Scholar ]
- 25. Meyer TF, Hill SA. In: Craig A, Scherf A, editors. Antigenic variation. Academic Press; 2003. Genetic variation in the pathogenic Neisseria spp. pp. 142–164. [ Google Scholar ]
- 26. Hill SA, Davies JK. Pilin gene variation in Neisseria gonorrhoeae: reassessing the old paradigms. FEMS Microbiol Rev. 2009;33(3):521–530. doi: 10.1111/j.1574-6976.2009.00171.x. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 27. Jonsson AB, Nyberg G, Normark S. Phase variation of gonococcal pili by frameshift mutation in pilC, a novel gene for pilus assembly. EMBO J. 1991;10(2):477–488. doi: 10.1002/j.1460-2075.1991.tb07970.x. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 28. Tonjum T, Koomey M. The pilus colonization factor of pathogenic neisserial species: organelle biogenesis and structure/function relationships- a review. Gene. 1997;192(1):155–163. doi: 10.1016/S0378-1119(97)00018-8. [ DOI ] [ PubMed ] [ Google Scholar ]
- 29. Fussenegger M, Rudel T, Barten R, Ryll R, Meyer TF. Transformation competence and type-4 pilus biogenesis in Neisseria gonorrhoeae- a review. Gene. 1997;192(1):125–134. doi: 10.1016/S0378-1119(97)00038-3. [ DOI ] [ PubMed ] [ Google Scholar ]
- 30. Wolfgang M, van Putten JP, Hayes SF, Dorward D, Koomey M. Components and dynamics of fiber formation define a ubiquitous biogenesis pathway for bacterial pili. EMBO J. 2000;19(23):6408–6418. doi: 10.1093/emboj/19.23.6408. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 31. Drake SL, Koomey M. The product of the pilQ gene is essential for the biogenesis of type IV pili in Neisseria gonorrhoeae. Mol Microbiol. 1995;18(5):975–986. doi: 10.1111/j.1365-2958.1995.18050975.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 32. Rudel T, Boxberger H-J, Meyer JF. Pilus biogenesis and epithelial cell adherence of Neisseria gonorrhoeae pilC double knock-out mutants. Mol Microbiol. 1995;17(6):1057–1071. doi: 10.1111/j.1365-2958.1995.mmi_17061057.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 33. Heckels JE. Structure and function of pili of pathogenic Neisseria species. Clin Microbiol Rev. 1989;2:S66–S73. doi: 10.1128/cmr.2.suppl.s66. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 34. Goodman SD, Scocca JJ. Identification and arrangement of the DNA sequences recognized in specific transformation of Neisseria gonorrhoeae. Proc Natl Acad Aci U S A. 1988;85(18):6982–6986. doi: 10.1073/pnas.85.18.6982. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 35. Koomey M, Gotschlich EC, Robbins K, Bergstrom S, Swanson J. Effects of recA mutations on pilus antigenic variation and phase transitions in Neisseria gonorrhoeae. Genetics. 1987;117(3):391–398. doi: 10.1093/genetics/117.3.391. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 36. Wolfgang M, van Putten JP, Hayes SF, Koomey M. The comP locus of Neisseria gonorrhoeae encodes a type IV prepilin that is dispensable for pilus biogenesis but essential for natural transformation. Mol Microbiol. 1999;31(5):1345–1357. doi: 10.1046/j.1365-2958.1999.01269.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 37. Aas FE, Wolfgang M, Frye S, Dunham S, Lovold C, Koomey M. Competence for natural transformation in Neisseria gonorrhoeae: components of DNA binding and uptake linked to type IV pilus expression. Mol Microbiol. 2002;46(3):749–760. doi: 10.1046/j.1365-2958.2002.03193.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 38. Cehovin A, Simpson PJ, McDowell MA, Brown DR, Noschese R, Pallett M, Brady J, Baldwin GS, Lea SM, Matthews SJ, Pelicic V. Specific DNA recognition mediated by a type IV pilin. Proc Natl Acad Aci U S A. 2013;110(8):3065–3070. doi: 10.1073/pnas.1218832110. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 39. Wolfgang M, Lauer P, Park H-S, Brossay L, Hebert J, Koomey M. PilT mutations lead to simultaneous defects in competence for natural transformation and twitching motility in piliated Neisseria gonorrhoeae. Mol Microbiol. 1998;29(1):321–330. doi: 10.1046/j.1365-2958.1998.00935.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 40. Maier B, Koomey M, Sheetz MP. A force-dependent switch reverses type IV pilus retraction. Proc Natl Acad Aci U S A. 2004;101(30):10961–10966. doi: 10.1073/pnas.0402305101. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 41. Johnston KH, Holmes KK, Gotschlich EC. The serological classification of Neisseria gonorrhoeae. I. Isolation of the Outer Membrane Complex Responsible for Serotypic Specificity. . J Exp Med. 1976;143(4):741–758. doi: 10.1084/jem.143.4.741. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 42. Sandstrom EG, Chen KC, Buchanan TM. Serology of Neisseria gonorrhoeae: coagglutination serotypes WI and WII/WIII correspond to different outer membrane protein I molecules. Infect Immun. 1982;38(2):462–470. doi: 10.1128/iai.38.2.462-470.1982. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 43. Cannon JG, Buchanan TM, Sparling PF. Confirmation of association of protein I serotype of Neisseria gonorrhoeae with the ability to cause disseminated infection. Infect Immun. 1983;40(2):816–819. doi: 10.1128/iai.40.2.816-819.1983. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 44. Judd RC. Protein I: structure, function and genetics. Clin Microbiol Rev. 1989;2:S41–S48. doi: 10.1128/cmr.2.suppl.s41. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 45. Lynch EC, Blake MS, Gotschlich EC, Mauro A. Studies of porins: Spontaneously transferred from whole cells and reconctituted from purified proteins of Neisseria gonorrhoeae and Neisseria meningitidis. Biophys J. 1984;45(1):104–107. doi: 10.1016/S0006-3495(84)84127-2. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 46. Weel JF, Hopman CT, van Putten JP. Bacterial entry and intracellular processing of Neisseria gonorrhoeae in epithelial cells: immunomorphological evidence for alterations in the major outer membrane protein P. IB. . Exp Med. 1991;174(3):705–715. doi: 10.1084/jem.174.3.705. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 47. Muller A, Rassow J, Grimm J, Machuy N, Meyer TF, Rudel T. VDAC and bacterial porin PorB of Neisseria gonorrhoeae share mitochondrial import pathways. EMBO J. 2002;21(8):1916–1929. doi: 10.1093/emboj/21.8.1916. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 48. Muller A, Gunther D, Dux F, Naumann M, Meyer TF, Rudel T. Neisserial porin (PorB) causes rapid calciuminflux in target cells and induces apoptosis by the activation of cysteine proteases. EMBO J. 1999;18(2):339–352. doi: 10.1093/emboj/18.2.339. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 49. Massari P, Ho Y, Wetzler LM. Neisseria meningitidis porin PorB interacts with mitochondria and protects cells from apoptosis. Proc Natl Acad Aci U S A. 2000;97(16):9070–9075. doi: 10.1073/pnas.97.16.9070. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 50. Rudel T, Schmid A, Benz R, Kolb H-A, Lang F, Meyer TF. Modulation of Neisseria porin (PorB) by cytosolic ATP/GTP of target cells: parallels between pathogen accommodation and mitochondrial endosymbiosis. Cell. 1996;85(3):391–402. doi: 10.1016/S0092-8674(00)81117-4. [ DOI ] [ PubMed ] [ Google Scholar ]
- 51. Mosleh IM, Huber LA, Steinlein P, Pasquali C, Gunther D, Meyer TF. Neisseria gonorrhoeae porin modulates phagosome maturation. J Biol Chem. 1998;273:35332–35338. doi: 10.1074/jbc.273.52.35332. [ DOI ] [ PubMed ] [ Google Scholar ]
- 52. Swanson J. Studies on gonococcus infection. XII. Colony color and opacity variants of gonococci. . Infect Immun. 1978;19(1):320–331. doi: 10.1128/iai.19.1.320-331.1978. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 53. Swanson J. Studies on gonococcus infection. XIV. Cell wall protein differences among color/opacity colony variants of gonococci. . Infect Immun. 1978;21(1):292–302. doi: 10.1128/iai.21.1.292-302.1978. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 54. Swanson J. Colony opacity and protein II compositions of gonococci. Infect Immun. 1982;37(1):359–368. doi: 10.1128/iai.37.1.359-368.1982. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 55. Bhat KS, Gibbs CP, Barrera O, Morrison SG, Jahnig F, Stern A, Kupsch E-M, Meyer TF, Swanson J. The opacity proteins of Neisseria gonorrhoeae strain MS11 are encoded by a family of 11 complete genes. Mol Microbiol. 1991;5(8):1889–1901. doi: 10.1111/j.1365-2958.1991.tb00813.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 56. Hauck CR, Meyer TF. "Small" talk: Opa proteins as mediators of Neisseria-host-cell communication. Curr Opin Microbiol. 2003;6(1):43–49. doi: 10.1016/S1369-5274(03)00004-3. [ DOI ] [ PubMed ] [ Google Scholar ]
- 57. Stern A, Brown M, Nickel P, Meyer TF. Opacity genes in Neisseria gonorrhoeae: control of phase and antigenic variation. Cell. 1986;47(1):61–71. doi: 10.1016/0092-8674(86)90366-1. [ DOI ] [ PubMed ] [ Google Scholar ]
- 58. Swanson J, Belland RJ, Hill SA. Neisserial surface variation: how and why? Curr Opin Genet Dev. 1992;2(5):805–811. doi: 10.1016/S0959-437X(05)80143-1. [ DOI ] [ PubMed ] [ Google Scholar ]
- 59. Swanson J, Barrera O, Sola J, Boslego J. Expression of outer membrane protein II by gonococci in experimental gonorrhea. J Exp Med. 1988;168(6):2121–2129. doi: 10.1084/jem.168.6.2121. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 60. Jerse AE, Cohen MS, Drown PM, Whicker LG, Isbey SF, Seifert HS, Cannon JG. Multiple gonococcal opacity proteins are expressed during experimental urethral infection in the male. J Exp Med. 1994;179(3):911–920. doi: 10.1084/jem.179.3.911. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 61. Makino S, van Putten JP, Meyer TF. Phase variation of the opacity outer membrane protein controls invasion by Neisseria gonorrhoeae into human epithelial cells. EMBO J. 1991;10(6):1307–1315. doi: 10.1002/j.1460-2075.1991.tb07649.x. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 62. van Putten JP, Paul SM. Binding of syndecan-like cell surface proteoglycan receptors is required for Neisseria gonorrhoeae entry into human mucosal cells. EMBO J. 1995;14(10):2144–2154. doi: 10.1002/j.1460-2075.1995.tb07208.x. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 63. Chen T, Belland RJ, Wilson J, Swanson J. Adherence of pilus minus Opa+ gonococci to epithelial cells in vitro involves heparin sulfate. J Exp Med. 1995;182(2):511–517. doi: 10.1084/jem.182.2.511. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 64. Chen T, Gotschlich EC. CGM1a antigen of neutrophils, a receptor for gonococcal opacity proteins. Proc Natl Acad Aci U S A. 1996;93(25):14851–14856. doi: 10.1073/pnas.93.25.14851. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 65. Virji M, Makepeace K, Ferguson DJ, Watt SM. Carcinoembryonic antigens (CD66) on epithelial cells and neutrophils are receptors for Opa proteins and pathogenic Neisseriae. Mol Microbiol. 1996;22(5):941–950. doi: 10.1046/j.1365-2958.1996.01551.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 66. Chen T, Grunert F, Medina-Marino A, Gotschlich EC. Several carcinoembryonic antigens (CD66) serve as receptors of gonococcal opacity proteins. J Exp Med. 1997;185(9):1557–1564. doi: 10.1084/jem.185.9.1557. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 67. Gray-Owen SD, Dehio C, Haude A, Grunert F, Meyer TF. CD66 carcinoembryonic antigens mediate interactions between Opa-expressing Neisseria gonorrhoeae and human polymorphonuclear phagocytes. EMBO J. 1997;16(12):3435–3445. doi: 10.1093/emboj/16.12.3435. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 68. Billker O, Popp A, Brinkmann V, Wenig G, Schneider J, Caron E, Meyer TF. Distinct mechanisms of internalization of Neisseria gonorrhoeae by members of the CEACAM receptor family involving Rac1- and Cdc42-dependent and -independent pathways. EMBO J. 2002;21(4):560–571. doi: 10.1093/emboj/21.4.560. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 69. Rice PA, Kasper DL. Characterization of gonococcal antigens responsible for induction of bacteriocidal antibody in disseminated infection: the role of gonococcal endotoxin. J Clin Invest. 1977;60(5):1149–1158. doi: 10.1172/JCI108867. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 70. Hook EWI, Olsen DA, Buchanan TM. Analysis of the antigen specificity of the human serum IgG immune response to complicated gonococcal infection. Infect Immun. 1984;43(2):706–709. doi: 10.1128/iai.43.2.706-709.1984. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 71. Apicella MA, Westerink MA, Morse SA, Schneider H, Rice PA, Griffiss JM. Bactericidal antibody response of normal human serum to the lipooligosaccharide of Neisseria gonorrhoeae. J Infect Dis. 1986;153(3):520–526. doi: 10.1093/infdis/153.3.520. [ DOI ] [ PubMed ] [ Google Scholar ]
- 72. Gregg CR, Melly MA, Hellergvist CG, Coniglio JG, McGee ZA. Toxic activity of purified lipopolysaccharide of Neisseria gonorrhoeae for human fallopian tube mucosa. J Infect Dis. 1981;143(3):432–439. doi: 10.1093/infdis/143.3.432. [ DOI ] [ PubMed ] [ Google Scholar ]
- 73. Schneider H, Griffiss JM, Boslego JW, Hitchcock PJ, Zahos KM, Apicella MA. Expression of paragloboside-like lipooligosaccharides may be a necessary component of gonococcal pathogenesis in men. J Exp Med. 1991;174(6):1601–1605. doi: 10.1084/jem.174.6.1601. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 74. van Putten JP, Robertson BD. Molecular mechanisms and implications for infection of lipopolysaccharide variationn in Neisseria. Mol Microbiol. 1995;16(5):847–853. doi: 10.1111/j.1365-2958.1995.tb02312.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 75. Danaher RJ, Levin JC, Arking D, Burch CL, Sandlin R, Stein DC. Genetic basis of Neisseria gonorrhoeae lipooligosaccharide antigenic variation. J Bacteriol. 1995;177(24):7275–7279. doi: 10.1128/jb.177.24.7275-7279.1995. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 76. Yang QL, Gotschlich EC. Variation of gonococcal lipopolysaccharide structure is due to alterations in poly-G tracts in lgt genes encoding glycosyl transferases. J Exp Med. 1996;183(1):323–327. doi: 10.1084/jem.183.1.323. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 77. Winstanley FP, Blackwell CC, Tan EL, Patel PV, Parsons NJ, Martin PM, Smith H. Alteration of pyocin-sensitivity pattern of Neisseria gonorrhoeae is associated with induced resistance to killing by human serum. Microbiology. 1984;130(5):1303–1306. doi: 10.1099/00221287-130-5-1303. [ DOI ] [ PubMed ] [ Google Scholar ]
- 78. Parsons NJ, Andrade JR, Patel PV, Cole JA, Smith H. Sialylation of lipopolysaccharide and loss of absorption of bactericidal antibody during conversion of gonococci to serum resistance by cytidine 5'-monphospho-N-acetyl neuraminic acid. Microbiol Pathog. 1989;7(1):63–72. doi: 10.1016/0882-4010(89)90112-5. [ DOI ] [ PubMed ] [ Google Scholar ]
- 79. Mandrell RE, Griffiss JM, Smith H, Cole JA. Distribution of of a lipopolysaccharide-specific sialyltransferase in pathogenic and non-pathogenic Neisseria. Microbiol Pathog. 1993;14(4):315–327. doi: 10.1006/mpat.1993.1031. [ DOI ] [ PubMed ] [ Google Scholar ]
- 80. de la Paz H, Cooke SJ, Heckels JE. Effect of sialylation of lipopolysaccharide of Neisseria gonorrhoeae on recognition and complemented-mediated killing by monoclonal antibodies directed against different outer-membrane antigens. Microbiology. 1995;141:913–920. doi: 10.1099/13500872-141-4-913. [ DOI ] [ PubMed ] [ Google Scholar ]
- 81. Elkins C, Carbonetti NH, Varela VA, Stirewalt D, Klapper DG, Sparling PF. Antibodies to N-terminal peptides of gonococcal porin are bacteriocidal when gonococcal lipopolysaccharide is not sialylated. Mol Microbiol. 1992;6(18):2617–2628. doi: 10.1111/j.1365-2958.1992.tb01439.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 82. van Putten JP. Phase variation of lipopolysaccharide directs interconversion of invasive and immune-resistant phenotypes of Neisseria gonorrhoeae. EMBO J. 1993;12(11):4043–4051. doi: 10.1002/j.1460-2075.1993.tb06088.x. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 83. Halter R, Pohlner J, Meyer TF. Mosaic-like organization of IgA protease genes in Neisseria gonorrhoeae generated by horizontal genetic exchange in vivo. EMBO J. 1989;8(9):2737–2744. doi: 10.1002/j.1460-2075.1989.tb08415.x. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 84. Pohlner J, Halter R, Beyreuther K, Meyer TF. Gene structure and extracellular secretion of Neisseria gonorrhoeae IgA protease. Nature. 1987;325:458–462. doi: 10.1038/325458a0. [ DOI ] [ PubMed ] [ Google Scholar ]
- 85. Halter R, Pohlner J, Meyer TF. IgA protease of Neisseria gonorrhoeae: isolation and characterization of the gene and its extracellular product. EMBO J. 1984;3(7):1595–15601. doi: 10.1002/j.1460-2075.1984.tb02016.x. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 86. Ayala P, Vasquez B, Wetzler L, So M. Neisseria gonorrhoeae porin P1. induces endosome exocytosis and redistribution of Lamp1 to the plasma membrane. . Infect Immun. 2002;70(11):5965–5971. doi: 10.1128/IAI.70.11.5965-5971.2002. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 87. Hopper S, Basquez B, Merz A, Clary S, Wilbur S, So M. Effects of the immunoglobulin A1 protease on Neisseria gonorrhoeae trafficking across polarized T84 epithelial monolayers. Infect Immun. 2000;68(2):906–911. doi: 10.1128/IAI.68.2.906-911.2000. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 88. Cornelissen CN. Molecular pathogenesis of Neisseria gonorrhoeae. Front Microbiol. 2011;2:224–226. doi: 10.3389/fmicb.2011.00224. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 89. Nassif X, Pujol C, Morand P, Eugene E. Interactions of pathogenic Neisseria with host cells. Is it possible to assemble the puzzle? Mol Microbiol. 1999;32(6):1124–1132. doi: 10.1046/j.1365-2958.1999.01416.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 90. Griffiss JM, Lammel CJ, Wang J, Dekker NP, Brooks GF. Neisseria gonorrhoeae Coordinately Uses Pili and Opa To Activate HEC-1-B Cell Microvilli, Which Causes Engulfment of the Gonococci. Infect Immun. 1999;67(7):3469–3480. doi: 10.1128/iai.67.7.3469-3480.1999. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 91. Edwards JL, Apicella MA. The Molecular Mechanisms Used by Neisseria gonorrhoeae To Initiate Infection Differ between Men and Women. Clin Microbiol Rev. 2004;17(4):965–981. doi: 10.1128/CMR.17.4.965-981.2004. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 92. Kallstrom H, Liszewski MK, Atkinson JP, Jonsson AB. Membrane cofactor protein (MCP or CD46) is a cellular pilus receptor for pathogenic Neisseria. Mol Microbiol. 1997;25(4):639–647. doi: 10.1046/j.1365-2958.1997.4841857.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 93. Wang J, Gray-Owen SD, Knorre A, Meyer TF, Dehio C. Opa binding to cellular CD66 receptors mediates the transcellular traversal of Neisseria gonorrhoeae across polarized T84 epithelial cell monolayers. Mol Microbiol. 1998;30(3):657–671. doi: 10.1046/j.1365-2958.1998.01102.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 94. James JF, Swanson J. Studies on gonococcus infection. XIII. Occurance of color/opacity colonial variants in clinical cultures. . Infect Immun. 1978;19(1):332–340. doi: 10.1128/iai.19.1.332-340.1978. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 95. Swanson KV, Jarvis GA, Brooks GF, Barham BJ, Cooper MD, Griffiss JM. CEACAM is not necessary for Neisseria gonorrhoeae to adhere to and invade female genital epithelial cells. Cell Microbiol. 2001;3(10):681–691. doi: 10.1046/j.1462-5822.2001.00147.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 96. Sintsova A, Wong H, MacDonald KS, Kaul R, Virji M, Gray-Owen SD. Selection of CEACAM receptor-specific binding phenotype during Neisseria gonorrhoeae infection of the human genital tract. Infect Immun. 2015;83(4):1372–1383. doi: 10.1128/iai.03123-14. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 97. Cole JG, Fulcher NB, Jerse AE. Opacity Proteins Increase Neisseria gonorrhoeae Fitness in the Female Genital Tract Due to a Factor under Ovarian Control. Infect Immun. 2010;78(4):1629–1641. doi: 10.1128/IAI.00996-09. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 98. Dehio C, Gray-Owen SD, Meyer TF. The role of neisserial Opa proteins in interactions with host cells. Trends Microbiol. 1998;6:489–495. doi: 10.1016/S0966-842X(98)01365-1. [ DOI ] [ PubMed ] [ Google Scholar ]
- 99. King GJ, Swanson J. Studies on gonococcus infection. XV. Identification of surface proteins of Neisseria gonorrhoeae. . Infect Immun. 1978;21(2):575–584. doi: 10.1128/iai.21.2.575-584.1978. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 100. Rest FR, Fischer SH, Ingham ZZ, Jones JF. Interactions of Neisseria gonorrhoeae with human neutrophils: effects of serum and gonococcal opacity on phagocytic killing and chemiluminescence. Infect Immun. 1982;36(2):737–744. doi: 10.1128/iai.36.2.737-744.1982. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 101. Rest RF, Lee N, Bowden C. Stimulation of human leukocytes by protein II+ gonococci is mediated by lectin-like gonococcal components. Infect Immun. 1985;50(1):116–122. doi: 10.1128/iai.50.1.116-122.1985. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 102. Ramsey KH, Schneider H, Cross AS, Boslego JW, Hoover DL, Staley TL, Kuschner RA, Deal CD. Inflammatory cytokines produced in response to experimental human gonorrhea. J Infect Dis. 1995;172(1):186–191. doi: 10.1093/infdis/172.1.186. [ DOI ] [ PubMed ] [ Google Scholar ]
- 103. Fisette PL, Ram S, Andersen JM, Guo W, Ingalls RR. The Lip lipoprotein from Neisseria gonorrhoeae stimulates cytokine release and NF-kappa beta activation in epithelial cells in a Toll-like receptor 2-dependent manner. J Biol Chem. 2003;278(47):46252–46260. doi: 10.1074/jbc.M306587200. [ DOI ] [ PubMed ] [ Google Scholar ]
- 104. Makepeace BL, Watt PJ, Heckels JE, Christodoulides M. Interactions of Neisseria gonorrhoeae with mature human macrophage opacity proteins influence production of proinflammatory cytokines. Infect Immun. 2001;69(3):1909–1913. doi: 10.1128/IAI.69.3.1909-1913.2001. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 105. Kaparakis M, Turnbull L, Carneiro L, Firth S, Coleman HA, Parkington HC, Le Bourhis L, Karrar A, Viala J, Mak J, Hutton ML, Davies JK, Crack PJ, Hertzog PJ, Philpott DJ, Girardin SE, Whitchurch CB, Ferrero RL. Bacterial membrane vesicles deliver peptidoglycan to NOP1 in epithelial cells. Cell Microbiol. 2010;12(3):372–385. doi: 10.1111/j.1462-5822.2009.01404.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 106. Ovcinnikov NM, Delektorskij VV. Electron microscope studies of gonococci in the urethral secretions of patients with gonorrhoea. Br J Vener Dis. 1971;47(6):419–439. doi: 10.1136/sti.47.6.419. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 107. 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(3):312–321. doi: 10.1038/mi.2009.139. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 108. Seib KL, Simons MP, Wu HJ, McEwan AG, Nauseef WM, Apicella MA, Jennings MP. Investigation of oxidative stress defenses of Neisseria gonorrhoeae by using a human polymorphonuclear leukocyte survival assay. Infect Immun. 2005;73(8):5269–5272. doi: 10.1128/IAI.73.8.5269-5272.2005. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 109. Criss AK, Seifert HS. A bacterial siren song: intimate interactions between Neisseria and neutrophils. Nat Rev Microbiol. 2012;10(3):178–190. doi: 10.1038/nrmicro2713. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 110. Seib KL, Wu HJ, Apicella MA, Jennings MP, McEwan AG. Defenses against oxidative stress in Neisseria gonorrhoeae: a system tailored for a challenging environment. Microbiol Mol Biol Rev. 2006;70(2):344–361. doi: 10.1128/MMBR.00044-05. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 111. Hill SA. In: Kidd S, editor. Stress Response in Pathogenic Bacteria. Advances in Molecular and Cellular Microbiology (Book 19) Cambridge: Cambridge University Press; 2011. Stress responses in pathogenic Neisseria: overlapping regulons and sRNA regulation. pp. 115–132. [ Google Scholar ]
- 112. 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(2):520–532. doi: 10.1111/j.1365-2958.2005.04839.x. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 113. Doherty CP. Host-pathogen interactions: the role of iron. J Nutr. 2007;137(5):1341–1344. doi: 10.1093/jn/137.5.1341. [ DOI ] [ PubMed ] [ Google Scholar ]
- 114. Jordan PW, Saunders NJ. Host iron binding proteins acting as niche indicators for Neisseria meningitidis. PLoS One. 2009;4(4):e5198. doi: 10.1371/journal.pone.0005198. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 115. Skaar EP. The battle for iron between bacterial pathogens and their vertebrate hosts. PLoS Pathog. 2010;6(8):e1000949. doi: 10.1371/journal.ppat.1000949. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 116. Noinaj N, Buchanan SK, Cornelissen CN. The transferrin-iron import system from pathogenic Neisseria species. Mol Microbiol. 2012;86(2):246–257. doi: 10.1111/mmi.12002. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 117. Noto JM, Cornelissen CN. Identification of TbpA residues required for transferrin-iron utilization by Neisseria gonorrhoeae. Infect Immun. 2008;76(5):1960–1969. doi: 10.1128/IAI.00020-08. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 118. Cornelissen CN, Biswas GD, Tsai J, Paruchuri DK, Thompson SA, Sparling PF. Gonococcal transferrin-binding protein 1 is required for transferrin utilization and is homologous to TonB-dependent outer membrane receptors. J Bacteriol. 1992;174(18):5788–5797. doi: 10.1128/jb.174.18.5788-5797.1992. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 119. Cornelissen CN, Kelley M, Hobbs MM, Anderson JE, Cannon JG, Cohen MS, Sparling PF. The transferrin receptor expressed by gonococcal strain FA1090 is required for the experimental infection of human male volunteers. Mol Microbiol. 1998;27(3):611–616. doi: 10.1046/j.1365-2958.1998.00710.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 120. Kasper DL, Rice PA, McCormick WM. Bacteriocidal antibody in genital infection due to Neisseria gonorrhoeae. J Infect Dis. 1977;135(2):243–251. doi: 10.1093/infdis/135.2.243. [ DOI ] [ PubMed ] [ Google Scholar ]
- 121. Ward ME, Lambden PR, Heckels JE, Watt PJ. The surface properties of Neisseria gonorrhoeae: determinants of susceptibility to antibody complement killing. J Gen Microbiol. 1978;108(2):205–212. doi: 10.1099/00221287-108-2-205. [ DOI ] [ PubMed ] [ Google Scholar ]
- 122. Schoolnik GK, Buchanan TM, Holmes KK. Gonococci causing disseminated gonococcl infection are resistant to the bactericidal action of normal human sera. J Clin Invest. 1976;58(5):1163–1173. doi: 10.1172/JCI108569. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 123. Joiner KA, Warren KA, Brown EJ, Swanson J, Frank MM. Studies on the mechanism of bacterial resistance to complement mediated killing. IV. C5b-9 forms high molecular weight complexes with bacterial outer membrane constituents on serum resistant but not on serum sensitive Neisseria gonorrhoeae. . J Immunol. 1983;131(3):1443–1451. [ PubMed ] [ Google Scholar ]
- 124. Joiner KA, Warren KA, Hammer C, Frank MM. Bactericidal but not nonbactericidal C5b-9 is associated with distinctive outer membrane proteins in Neisseria gonorrhoeae. J Immunol. 1985;134(3):1920–1925. [ PubMed ] [ Google Scholar ]
- 125. Lewis LA, Shafer WM, Dutta Ray T, Ram S, Rice PA. Phosphoethanolamine residues on the lipid A moiety of Neisseria gonorrhoeae lipooligosaccharide modulate binding of complement inhibitors and reistance to complement killing. Infect Immun. 2013;81(1):33–42. doi: 10.1128/IAI.00751-12. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 126. Joiner KA, Scales R, Warren KA, Frank MM, Rice PA. Mechanism of action of blocking immunoglobulin G for Neisseria gonorrhoeae. J Clin Invest. 1985;76(5):1765–1772. doi: 10.1172/JCI112167. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 127. Brooks GF, Lammel CJ, Petersen BH, Stites DP. Human seminal plasma inhibition of antibody complement-mediated killing and opsonisation of Neisseria gonorrhoeae and other Gram negative organisms. J Clin Invest. 1981;67(5):1523–1531. doi: 10.1172/JCI110183. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 128. Ram S, McQuillen DP, Gulati S, Elkins C, Pangburn MK, Rice PA. Binding of complement factor H to loop 5 of porin protein 1A: a molecular mechanism of serum resistance of nonsialylated Neisseria gonorrhoeae. J Exp Med. 1998;188(4):671–680. doi: 10.1084/jem.188.4.671. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 129. Ngampasutadol J, Ram S, Gulati S, Agarwal S, Li C, Visintin A, Monks B, Madico G, Rice PA. Human factor H interacts selectively with Neisseria gonorrhoeae and results in species-specific complement evasion. J Immunol. 2008;180(5):3426–3435. doi: 10.4049/jimmunol.180.5.3426. [ DOI ] [ PubMed ] [ Google Scholar ]
- 130. Ngampasutadol J, Ram S, Blom AM, Jarva H, Jerse AE, Lien E, Goguen J, Gulati S, Rice PA. Human C4b-binding protein selectively interacts with Neisseria gonorrhoeae and results in species-specific infection. Proc Natl Acad Aci U S A. 2005;102(47):17142–17147. doi: 10.1073/pnas.0506471102. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 131. Blom A, Ram S. Contribution of interactions between complement inhibitor C4b-binding protein and pathogens to their ability to establish infection with particular emphasis on Neisseria gonorrhoeae. Vaccine. 2008;26(Suppl 8):149–155. doi: 10.1016/j.vaccine.2008.11.049. [ DOI ] [ PubMed ] [ Google Scholar ]
- 132. Tramont EC. Inhibition of adherence of Neisseria gonorrhoeae by human genital secretions. J Clin Invest. 1977;59(1):117–124. doi: 10.1172/JCI108608. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 133. Hedges SR, Mayo MS, Mestecky J, Hook EW 3rd, Russell MW. Limited local and systemic antibody responses to Neisseria gonorrhoeae during uncomplicated genital infections. Infect Immun. 1999;67(8):3937–3946. doi: 10.1128/iai.67.8.3937-3946.1999. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 134. Boulton IC, Gray-Owen SD. Neisserial binding to CEACAM1 arrests the activation and proliferation of CD4+ T lymphocytes. Nat Immunol. 2002;3(3):229–236. doi: 10.1038/ni769. [ DOI ] [ PubMed ] [ Google Scholar ]
- 135. Lee HS, Ostrowski MA, Gray-Owen SD. CEACAM1 dynamics during Neisseria gonorrhoeae suppression of CD4+ T lymphocyte activation. J Immunol. 2008;180(10):6827–6835. doi: 10.4049/jimmunol.180.10.6827. [ DOI ] [ PubMed ] [ Google Scholar ]
- 136. Pantelic M, Kim YJ, Bolland S, Chen I, Shively J, Chen T. Neisseria gonorrhoeae kills carcinoembryonic antigen-related cellular adhesion molecule 1 (CD66a)-expressing human B cells and inhibits antibody production. Infect Immun. 2005;73(7):4171–4179. doi: 10.1128/IAI.73.7.4171-4179.2005. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 137. Zhang P, Schwartz O, Pantelic M, Li G, Knazze Q, Nobile C, Radovich M, He J, Hong SC, Klena J, Chen T. DC-SIGN (CD209) recognition of Neisseria gonorrhoeae is circumvented by lipooligosaccharide variation. J Leukoc Biol. 2006;79(4):731–738. doi: 10.1189/jlb.0405184. [ DOI ] [ PubMed ] [ Google Scholar ]
- 138. Lu M, Zhang M, Takashima A, Weiss J, Apicella MA, Li XH, Yuan D, Munford RS. Lipopolysaccharide deacylation by an endogenous lipase controls innate antibody responses to Gram-negative bacteria. Nat Immunol. 2005;6(10):989–994. doi: 10.1038/ni1246. [ DOI ] [ PubMed ] [ Google Scholar ]
- 139. Jerse AE. Experimental gonococcal genital tract infection and opacity protein expression in estradiol-treated mice. Infect Immun. 1999;67(11):5699–5708. doi: 10.1128/iai.67.11.5699-5708.1999. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 140. Jerse AE, Wu H, Packiam M, Vonck RA, Begum AA, Garvin LE. 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 ]
- 141. 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(3):e00095–e00011. doi: 10.1128/mBio.00095-11. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 142. Liu Y, Islam EA, Jarvis GA, Gray-Owen SD, Russell MW. Neisseria gonorrhoeae selectively suppresses the devolpment of Th1 and Th2 cells, and enhances Th17 cell responses, through TGF-beta-dependent mechanisms. Mucosal Immunol. 2012;5(3):320–331. doi: 10.1038/mi.2012.12. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 143. So NS, Ostrowski MA, Gray-Owen SD. Vigorous response of human innate functioning IgM memory B cells upon infection by Neisseria gonorrhoeae. J Immunol. 2012;188(8):4008–4022. doi: 10.4049/jimmunol.1100718. [ DOI ] [ PubMed ] [ Google Scholar ]
- 144. Muenzner P, Bachmann V, Zimmermann W, Hentschel J, Hauck CR. Human-restricted bacterial pathogens block shedding of epithelial cells by stimulating integrin activation. Science. 2010;329(5996):1197–1201. doi: 10.1126/science.1190892. [ DOI ] [ PubMed ] [ Google Scholar ]
- 145. Li G, Jiao H, Yan H, Wang J, Wang X, Ji M. Establishment of human CEACAM1 transgenic mouse model for the study of gonococcal infections. J Microbiol Methods. 2011;87(3):350–354. doi: 10.1016/j.mimet.2011.09.012. [ DOI ] [ PubMed ] [ Google Scholar ]
- 146. Snyder LA, Butcher SA, Saunders NJ. Comparative whole-genome analyses reveal over 100 putative phase-variable genes in the pathogenic Neisseria spp. Microbiology. 2001;147(Pt 8):2321–2332. doi: 10.1099/00221287-147-8-2321. [ DOI ] [ PubMed ] [ Google Scholar ]
- 147. Moxon R, Bayliss C, Hood D. Bacterial contingency loci: the role of simple sequence DNA repeats in bacterial adaptation. Annu Rev Genet. 2006;40:307–333. doi: 10.1146/annurev.genet.40.110405.090442. [ DOI ] [ PubMed ] [ Google Scholar ]
- 148. Haas R, Veit S, Meyer TF. Silent pilin genes of Neisseria gonorrhoeae MS11 and the occurance of related hypervariant sequences among other gonococcal isolates. Mol Microbiol. 1992;6(2):197–208. doi: 10.1111/j.1365-2958.1992.tb02001.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 149. Anderson MT, Dewenter L, Maier B, Seifert HS. Seminal plasma initiates a Neisseria gonorrhoeae transmission state. Mol Microbiol. 2014;5(2):e01004–e01013. doi: 10.1128/mBio.01004-13. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 150. Sparling PF, Elkins C, Wyrick PB, Cohen MS. Vaccines for bacterial sexually transmitted infections: a realistic goal? Proc Natl Acad Aci U S A. 1994;91(7):2456–2463. doi: 10.1073/pnas.91.7.2456. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 151. Russell MW, Hedges SR, Wu HY, Hook EW 3rd, Mestecky J. Mucosal immunity in the genital tract: prospects for vaccines against sexually transmitted diseases - a review. Am J Reprod Immunol. 1999;42(1):58–63. doi: 10.1111/j.1600-0897.1999.tb00466.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 152. Jerse AE, Bash MC, Russell MW. Vaccines against gonorrhea: current status and future challenges. Vaccine. 2014;32(14):1579–1587. doi: 10.1016/j.vaccine.2013.08.067. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 153. McChesney D, Tramont EC, Boslego JW, Ciak J, Sadoff J, Brinton CC. Genital antibody response to a parenteral gonococcal pilus vaccine. Infect Immun. 1982;36(3):1006–1012. doi: 10.1128/iai.36.3.1006-1012.1982. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 154. Tramont EC, Boslego JW. Pilus vaccines. Vaccine. 1985;3(1):3–10. doi: 10.1016/0264-410X(85)90003-9. [ DOI ] [ PubMed ] [ Google Scholar ]
- 155. Simpson SD, Ho Y, Rice PA, Wetzler LM. T lymphocyte response to Neisseria gonorrhoeae porin in individuals with mucosal gonococcal infections. J Infect Dis. 1999;180(3):762–773. doi: 10.1086/314969. [ DOI ] [ PubMed ] [ Google Scholar ]
- 156. Massari P, Ram S, Macleod H, Wetzler LM. The role porins in neisserial pathogenesis and immunity. Trends Microbiol. 2003;11(2):87–93. doi: 10.1016/S0966-842X(02)00037-9. [ DOI ] [ PubMed ] [ Google Scholar ]
- 157. Singleton TE, Massari P, Wetzler LM. Neisserial porin-induced dendritic cell activation is MyD88 and TLR2 dependent. J Immunol. 2005;174(6):3545–3550. doi: 10.4049/jimmunol.174.6.3545. [ DOI ] [ PubMed ] [ Google Scholar ]
- 158. Zhu W, Thomas CE, Chen CJ, Van Dam CN, Johnston RE, Davis NL, Sparling PF. Comparison of immune responses to gonococcal PorB delivered as outer membrane vesicles, recombinant protein, or Venezuelan equine encephalitis virus replicon particles. Infect Immun. 2005;73(11):7558–7568. doi: 10.1128/IAI.73.11.7558-7568.2005. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 159. Liang Y, Hasturk H, Elliot J, Noronha A, Liu X, Wetzler LM, Massari P, Kantarci A, Winter HS, Farraye FA, Ganley-Leal LM. Toll-like receptor 2 induces mucosal homing receptor expression and IgA production by human B cells. Clin Immunol. 2011;138(1):33–40. doi: 10.1016/j.clim.2010.09.003. [ DOI ] [ PubMed ] [ Google Scholar ]
- 160. Rice PA, Kasper DL. Characterization of serum resistance of Neisseria gonorrhoeae that disseminate: roles of blocking antibody and gonococcal outer membrane proteins. J Clin Invest. 1982;70(1):157–167. doi: 10.1172/jci110589. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 161. Rice PA, McQuillen DP, Gulati S, Jani DB, Wetzler LM, Blake MS, Gotschlich EC. Serum resistance of Neisseria gonorrhoeae. Does it thwart the inflammatory response and facilitate transmission of infection? Ann NY Acad Sci. 1994;730(1):7–14. doi: 10.1111/j.1749-6632.1994.tb44234.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 162. Plummer FA, Chubb H, Simonsen JN, Bosire M, Slaney L, Maclean I, Ndinya-Achola JO, Waiyaki P, Brunham RC. Antibody to Rmp (outer membrane protein 3) increases susceptibility to gonococcal infection. J Clin Invest. 1993;91(1):339–343. doi: 10.1172/jci116190. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 163. Newhall WJ, Haak RA. Cross-linking analysis of the outer membrane proteins of Neisseria gonorrhoeae. Infect Immun. 1980;28(3):785–791. doi: 10.1128/iai.28.3.785-791.1980. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 164. Maiden MC, Bygraves JA, Feil E, Morelli G, Russell JE, Urwin R, Zhang Q, Zhou J, Zurth K, Caugant DA, Feavers IM, Achtman M, Spratt BG. Multilocus sequence typing: A portable approach to the identification of clones within populations of pathogenic microorganisms. Proc Natl Acad Aci U S A. 1998;95(6):3140–3145. doi: 10.1073/pnas.95.6.3140. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 165. Carifo K, Catlin BW. Neisseria gonorrhoeae Auxotyping: Differentiation of Clinical Isolates Based on Growth Responses on Chemically Defined Media. Appl Microbiol. 1973;26(3):223–230. doi: 10.1128/am.26.3.223-230.1973. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 166. La Scolea LJJ, Young FE. Development of a Defined Minimal Medium for the Growth of Neisseria gonorrhoeae. Appl Microbiol. 1974;28(1):70–76. doi: 10.1128/am.28.1.70-76.1974. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 167. Apicella MA, Shero M, Jarvis GA, Griffiss JM, Mandrell RE, Schneider H. Phenotypic variation in epitope expression of the Neisseria gonorrhoeae lipooligosaccharide. Infect Immun. 1987;55(8):1755–1761. doi: 10.1128/iai.55.8.1755-1761.1987. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 168. Knapp JS, Holmes KK. Disseminated Gonococcal Infections Caused by Neisseria gonorrhoeae with Unique Nutritional Requirements. J Infect Dis. 1975;132(2):204–208. doi: 10.1093/infdis/132.2.204. [ DOI ] [ PubMed ] [ Google Scholar ]
- 169. Knapp JS, Holmes KK, Bonin P, Hook EW 3rd. Epidemiology of Gonorrhea: Distribution and Temporal Changes in Auxotype/Serovar Classes of Neisseria gonorrhoeae. Sex Transm Dis. 1987;14(1):26–32. doi: 10.1097/00007435-198701000-00006. [ DOI ] [ PubMed ] [ Google Scholar ]
- 170. Knapp JS, Tam MR, Nowinski RC, Holmes KK, Sandstrom EG. Serological Classification of Neisseria gonorrhoeae with Use of Monoclonal Antibodies to Gonococcal Outer Membrane Protein I. J Infect Dis. 1984;150(1):44–48. doi: 10.1093/infdis/150.1.44. [ DOI ] [ PubMed ] [ Google Scholar ]
- 171. Buchanan TM, Hildebrandt JF. Antigen-Specific Serotyping of Neisseria gonorrhoeae: Characterization Based Upon Principal Outer Membrane Protein. Infect Immun. 1981;32(3):985–994. doi: 10.1128/iai.32.3.985-994.1981. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 172. Eisenstein BI, Lee TJ, Sparling PF. Penicillin Sensitivity and Serum Resistance are Independent Attributes of Strains of Neisseria gonorrhoeae Causing Disseminated Gonococcal Infection. Infect Immun. 1977;15(3):834–841. doi: 10.1128/iai.15.3.834-841.1977. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 173. Wiesner PJ, Handsfield HH, Holmes KK. Low antibiotic resistance of gonococci causing disseminated infection. N Engl J Med. 1973;288(23):1221–1222. doi: 10.1056/NEJM197306072882308. [ DOI ] [ PubMed ] [ Google Scholar ]
- 174. O’Brien JP, Goldenberg DL, Rice PA. Disseminated Gonococcal Infection: A Prospective Analysis of 49 Patients and a Review of Pathophysiology and Immune Mechanisms. Medicine. 1983;62(6):395–406. doi: 10.1097/00005792-198311000-00005. [ DOI ] [ PubMed ] [ Google Scholar ]
- 175. Sarubbi FAJ, Blackman E, Sparling PF. Genetic Mapping of Linked Antibiotic Resistance Loci in Neisseria gonorrhoeae. J Bacteriol. 1974;120(3):1284–1292. doi: 10.1128/jb.120.3.1284-1292.1974. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 176. Perine PL, Schalla W, Siegel MS, Thornsberry C, Biddle J, Wong K-H, Thompson SE. Evidence for Two Distinct Types of Penicillinase-producing Neisseria gonorrhoeae. Lancet. 1977;2(8046):993–995. doi: 10.1016/S0140-6736(77)92891-4. [ DOI ] [ PubMed ] [ Google Scholar ]
- 177. Handsfield KK, Sandström EG, Knapp JS, Perine PL, Whittington WL, Sayers DE, Holmes KK. Epidemiology of Penicillinase-Producing Neisseria gonorrhoeae Infections — Analysis by Auxotyping and Serogrouping. N Engl J Med. 1982;306:950–954. doi: 10.1056/NEJM198204223061602. [ DOI ] [ PubMed ] [ Google Scholar ]
- 178. Handsfield HH, Knapp JS, Diehr PK, Holmes KK. Correlation of Auxotype and Penicillin Susceptibility of Neisseria gonorrhoeae with Sexual Preference and Clinical Manifestations of Gonorrhea. Sex Transm Dis. 1980;7(1):1–5. doi: 10.1097/00007435-198001000-00001. [ DOI ] [ PubMed ] [ Google Scholar ]
- 179. Yorke JA, Hethcote HW, Nold A. Dynamics and Control of the Transmission of Gonorrhea. Sex Transm Dis. 1978;5:51–56. doi: 10.1097/00007435-197804000-00003. [ DOI ] [ PubMed ] [ Google Scholar ]
- 180. Handsfield HH, Rice RJ, Roberts MC, Holmes KK. Localized Outbreak of Penicillinase-Producing Neisseria gonorrhoeae: Paradigm for Introduction and Spread of Gonorrhea in a Community. J Amer Med Assoc. 1989;261(16):2357–2361. doi: 10.1001/jama.1989.03420160089028. [ DOI ] [ PubMed ] [ Google Scholar ]
- 181. Potterat JJ, Rothenberg R, Bross DC. Gonorrhea in Street Prostitutes: Epidemiological and Legal Implications. Sex Transm Dis. 1979;6(2):58–63. doi: 10.1097/00007435-197904000-00003. [ DOI ] [ PubMed ] [ Google Scholar ]
- 182. Upchurch DM, Brady WE, Reichart CA, Hook EW 3rd. Behavioral Contributions to Acquisition of Gonorrhea in Patients Attending an Inner City Sexually Transmitted Disease Clinic. J Infect Dis . 1990;161(5):938–941. doi: 10.1093/infdis/161.5.938. [ DOI ] [ PubMed ] [ Google Scholar ]
- 183. Platt R, Rice PA, McCormack WM. Risk of Acquiring Gonorrhea and Prevalence of Abnormal Adnexal Findings Among Women Recently Exposed to Gonorrhea. J Amer Med Assoc. 1983;250(23):3205–3209. doi: 10.1001/jama.1983.03340230057031. [ DOI ] [ PubMed ] [ Google Scholar ]
- 184. World Health Organziation, Switzerland. Neisseria gonorrhoeae and gonococcal infections. 1978 [ PubMed ] [ Google Scholar ]
- 185. Spratt BG, Maiden MC. Bacterial population genetics, evolution and epidemiology. Phil Trans Royal Soc B: Biol Sci. 1999;354(1384):701–710. doi: 10.1098/rstb.1999.0423. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 186. Bennett JS, Jolley KA, Sparling PF, Saunders NJ, Hart CA, Feavers IM, Maiden MC. Species status of Neisseria gonorrhoeae: evolutionary and epidemiological inferences from multilocus sequence typing. BMC Biology . 2007;5:35. doi: 10.1186/1741-7007-5-35. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 187. Selander RK, Beltran P, Smith NH, Barker RM, Crichton PB, Old DC, Musser JM, Whittam TS. Genetic population structure, clonal phylogeny, and pathogenicity of Salmonella paratyphi B. Infect Immun. 1990;58(6):1891–1901. doi: 10.1128/iai.58.6.1891-1901.1990. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 188. Selander RK, Caugant DA, Ochman H, Musser JM, Gilmour MN, Whittam TS. Methods of Multilocus Enzyme Electrophoresis for Bacterial Population Genetics and Systematics. App Environ Microbiol. 1986;51(5):873–884. doi: 10.1128/aem.51.5.873-884.1986. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 189. Maynard Smith J, Smith NH, O’Rourke M, Spratt BG. How clonal are bacteria? Proc Natl Acad Aci U S A. 1993;90(10):4384–4388. doi: 10.1073/pnas.90.10.4384. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 190. Selander RK, Levin BR. Genetic diversity and structure in Escherichia coli populations. Science. 1980;210(4469):545–547. doi: 10.1126/science.6999623. [ DOI ] [ PubMed ] [ Google Scholar ]
- 191. Caugant DA, Levin BR, Selander RK. Genetic Diversity and Temporal Variation in the E. coli Population of a Human Host. . Genetics. 1981;98(3):467–490. doi: 10.1093/genetics/98.3.467. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 192. Feil EJ, Li BC, Aanensen DM, Hanage WP, Spratt BG. eBURST: Inferring Patterns of Evolutionary Descent among Clusters of Related Bacterial Genotypes from Multilocus Sequence Typing Data. J Bacteriol. 2004;186(5):1518–1530. doi: 10.1128/JB.186.5.1518-1530.2004. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 193. Palmer HM, Arnold C. Genotyping Neisseria gonorrhoeae Using Fluorescent Amplified Fragment Length Polymorphism Analysis. J Clin Microbiol. 2001;39(6):2325–2329. doi: 10.1128/JCM.39.6.2325-2329.2001. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 194. Feil EJ, Spratt BG. Recombination and the population structures of bacterial pathogens. Ann Rev Microbiol. 2001;55:561–590. doi: 10.1146/annurev.micro.55.1.561. [ DOI ] [ PubMed ] [ Google Scholar ]
- 195. Bennett JS, Griffiths DT, McCarthy ND, Sleeman KL, Jolley KA, Crook DW, Maiden MC. Genetic Diversity and Carriage Dynamics of Neisseria lactamica in Infants. Infect Immun. 2005;73(4):2424–2432. doi: 10.1128/IAI.73.4.2424-2432.2005. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 196. Feil EJ, Holmes EC, Bessen DE, Chan M-S, Day NP, Enright MC, Goldstein R, Hood DW, Kalia A, Moore CE, Zhou J, Spratt BG. Recombination within natural populations of pathogenic bacteria: short-term empirical estimates and long-term phylogenetic consequences. Proc Natl Acad Aci U S A. 2001;98:182–187. doi: 10.1073/pnas.98.1.182. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 197. Jolley KA, Feil EJ, Chan M-S, Maiden MC. Sequence type analysis and recombinational tests (START). Bioinformatics. 2001;17(12):1230–1231. doi: 10.1093/bioinformatics/17.12.1230. [ DOI ] [ PubMed ] [ Google Scholar ]
- 198. Perez-Losada M, Crandall KA, Bash MC, Dan M, Zenilman J, Viscidi RP. Distinguishing importation from diversification of quinolone-resistant Neisseria gonorrhoeae by molecular evolutionary analysis. . BMC Evol Biol . 2007;7:84. doi: 10.1186/1471-2148-7-84. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 199. Johnson SR, Morse SA. Antibiotic Resistance in Neisseria gonorrhoeae: Genetics and Mechanisms of Resistance. Sex Transm Dis. 1988;15(4):217–224. doi: 10.1097/00007435-198810000-00008. [ DOI ] [ PubMed ] [ Google Scholar ]
- 200. Rohlfing SR, Landmesser JK, Gerster JF, Sharon EP, Stern RM. Differentiation of fluorinated quinolone antibacterials with Neisseria gonorrhoeae isolates. J Antimicrobial Chem. 1985;15(4):539–544. doi: 10.1093/jac/15.5.539. [ DOI ] [ PubMed ] [ Google Scholar ]
- 201. Macaulay ME. Acrosoxacin resistance in Neisseria gonorrhoeae. Lancet. 1982;319(8264):171–172. doi: 10.1016/s0140-6736(82)90423-8. [ DOI ] [ PubMed ] [ Google Scholar ]
- 202. Vidovic S, Thakur SD, Horsman GB, Levett PN, Anvari V, Dillon J-A. Longitudinal Analysis of the Evolution and Dissemination of Neisseria gonorrhoeae Strains (Saskatchewan, Canada, 2005 to 2008) Reveals Three Major Circulating Strains and Convergent Evolution of Ciprofloxacin and Azithromycin Resistance. J Clin Microbiol. 2012;50(12):3823–3830. doi: 10.1128/jcm.01402-12. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 203. Spratt BG, Hanage WP, Li B, Aanensen DM, Feil EJ. Displaying the relatedness among isolates of bacterial species - the eBURST approach. FEMS Microbiol Letts. 2004;241:129–134. doi: 10.1016/j.femsle.2004.11.015. [ DOI ] [ PubMed ] [ Google Scholar ]
- 204. Ohnishi M, Golparian D, Shimuta K, Saika T, Hoshina S, Iwasaku K, Nakayama S-I, Kitawaki J, Unemo M. Is Neisseria gonorrhoeae initiating a future era of untreatable gonorrhea? Detailed characterization of the first strain with high-level resistance to ceftriaxone . Antimicrob Agents Chemo. 2011;55(7):3538–3545. doi: 10.1128/aac.00325-11. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 205. Unemo M, Golparian D, Nicholas R, Ohnishi M, Gallay A, Sednaoui P. High-Level Cefixime- and Ceftriaxone-Resistant Neisseria gonorrhoeae in France: Novel penA Mosaic Allele in a Successful International Clone Causes Treatment Failure. Antimicrob Agents Chemo. 2011;56(3):1273–1280. doi: 10.1128/aac.05760-11. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 206. Camara J, Serra J, Ayats J, Bastida T, Carnicer-Pont D, Andreu A, Ardanuy C. Molecular characterization of two high-level ceftriaxone-resistant Neisseria gonorrhoeae isolates detected in Catalonia, Spain. J Antimicrobial Chem. 2012;67(8):1858–1860. doi: 10.1093/jac/dks162. [ DOI ] [ PubMed ] [ Google Scholar ]
- 207. Unemo M, Nicholas RA. Emergence of multidrug-resistant, extensively drug-resistant and untreatable gonorrhea. Future Microbiol. 2012;7(12):1401–1422. doi: 10.2217/fmb.12.117. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 208. Unemo M, Shafer WM. Antimicrobial resistance in Neisseria gonorrhoeae in the 21st century: past, evolution, and future. Clin Microbiol Rev. 2014;27(3):587–613. doi: 10.1128/cmr.00010-14. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 209. Unemo M, Shafer WM. Antibiotic resistance in Neisseria gonorrhoeae: origin, evolution, and lessons learned for the future. Ann NY Acad Sci. 2011;1230(1):E19–E28. doi: 10.1111/j.1749-6632.2011.06215.x. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 210. Tanaka M, Tunoe H, Egashira T, Naito S, Nakayama H, Kanayama A, Saika T, Kobayashi I. A remarkable reduction in the susceptibility of Neisseria gonorrhoeae isolates to cephems and the selection of antibiotic regimens for the single-dose treatment of gonococcal infection in Japan. J Infect Chemo. 2002;8(1):81–86. doi: 10.1007/s101560200011. [ DOI ] [ PubMed ] [ Google Scholar ]
- 211. Furuya R, Tanaka M, Onoye Y, Kanayama A, Saika T, Iyoda T, Tatewaki M, Matsuzaki K, Kobayashi I. Antimicrobial resistance in clinical isolates of Neisseria subflava from the oral cavities of a Japanese population. J Infect Chemo. 2007;13(5):302–304. doi: 10.1007/s10156-007-0541-8. [ DOI ] [ PubMed ] [ Google Scholar ]
- 212. Ashford WA, Golash RG, Henning VG. Penicillinase producing Neisseria gonorrhoeae. Lancet. 1976;308(7987):657–658. doi: 10.1016/s0140-6736(76)92467-3. [ DOI ] [ PubMed ] [ Google Scholar ]
- 213. Phillips I. Beta-lactamase producing penicillin-resistant gonococcus. Lancet. 1976;308(7987):656–657. doi: 10.1016/s0140-6736(76)92466-1. [ DOI ] [ PubMed ] [ Google Scholar ]
- 214. Cannon J, Sparling PF. The genetics of the gonococcus. Ann Rev Microbiol. 1984;38(1):111–133. doi: 10.1146/annurev.micro.38.1.111. [ DOI ] [ PubMed ] [ Google Scholar ]
- 215. Morse SA, Johnson SR, Biddle JW, Roberts MC. High-level tetracycline resistance in Neisseria gonorrhoeae is result of acquisition of streptococcal tetM determinant. Antimicrob Agents Chemo. 1986;30(5):664–670. doi: 10.1128/aac.30.5.664. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 216. Sox TE, Mohammed W, Blackman E, Biswas G, Sparling PF. Conjugative plasmids in Neisseria gonorrhoeae. J Bacteriol. 1978;134(1):278–286. doi: 10.1128/jb.134.1.278-286.1978. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 217. Eisenstein B, Sox T, Biswas G, Blackman E, Sparling P. Conjugal transfer of the gonococcal penicillinase plasmid. Science. 1977;195(4282):998–1000. doi: 10.1126/science.402693. [ DOI ] [ PubMed ] [ Google Scholar ]
- 218. Kirven LA, Thornsberry C. Transfer of beta-lactamase genes of Neisseria gonorrhoeae by conjugation. Antimicrob Agents Chemo. 1977;11(6):1004–1006. doi: 10.1128/aac.11.6.1004. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 219. Powell AJ, Tomberg J, Deacon AM, Nicholas RA, Davies C. Crystal structures of Por from penicillin-susceptible and -resistant strains of Neisseria gonorrhoeae reveal an unexpectedly subtle mechanism for antibiotic resistance. J Biol Chem. 2008;284(2):1202–1212. doi: 10.1074/jbc.m805761200. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 220. Sparling PF, Sarubbi FA, Blackman E. Inheritance of low-level resistance to penicillin, tetracycline, and chloramphenicol in Neisseria gonorrhoeae. J Bacteriol. 1975;124(2):740–749. doi: 10.1128/jb.124.2.740-749.1975. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 221. Zhao S, Duncan M, Tomberg J, Davies C, Unemo M, Nicholas RA. Genetics of chromosomally mediated intermediate resistance to ceftriaxone and cefixime in Neisseria gonorrhoeae. Antimicrob Agents Chemo. 2009;53(9):3744–3751. doi: 10.3410/f.1168418.630600. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 222. 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(2):462–478. doi: 10.1111/j.1365-2958.2008.06424.x. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 223. 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(12):1804–1812. doi: 10.1111/j.1365-2958.2008.06424.x. [ DOI ] [ PubMed ] [ Google Scholar ]
- 224. Veal WL, Nicholas RA, Shafer WM. Overexpression of the MtrC-MtrD-MtrE efflux pump due to an mtrR mutation is required for chromosomally mediated penicillin resistance in Neisseria gonorrhoeae. J Bacteriol. 2002;184(20):5619–5624. doi: 10.1128/jb.184.20.5619-5624.2002. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 225. Zarantonelli L, Borthagaray G, Lee EH, Shafer WM. Decreased azithromycin susceptibility of Neisseria gonorrhoeae due to mtrR mutations. Antimicrob Agents Chemo. 1999;43(10):2468–2472. doi: 10.1128/aac.43.10.2468. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 226. Golparian D, Shafer WM, Ohnishi M, Unemo M. Importance of multidrug efflux pumps in the antimicrobial resistance property of clinical multidrug-resistant isolates of Neisseria gonorrhoeae. Antimicrob Agents Chemo. 2014;58(6):3556–3559. doi: 10.1128/aac.00038-14. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 227. Shafer WM, Qu X-D, Waring AJ, Lehrer RI. Modulation of Neisseria gonorrhoeae susceptibility to vertebrate antibacterial peptides due to a member of the resistance/nodulation/division efflux pump family. Proc Natl Acad Aci U S A. 1998;95(4):1829–1833. doi: 10.1073/pnas.95.4.1829. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 228. Bignell C, Unemo M. 2012 European guideline on the diagnosis and treatment of gonorrhoea in adults. Int J STD AIDS. 2013;24(2):85–92. doi: 10.1177/0956462412472837. [ DOI ] [ PubMed ] [ Google Scholar ]
- 229. Center for Disease Control and Prevention Update to CDC’s sexually transmitted diseases treatment guidelenes, 2010: Oral cephalosporins no longer a recommended treatment for gonococcal infections. JAMA . 2012;308(18):1850. doi: 10.1001/2012.jama.11292. [ DOI ] [ PubMed ] [ Google Scholar ]
- 230. Kirkcaldy RD, Weinstock HS, Moore PC, Philip SS, Wiesenfeld HC, Papp JR, Kerndt PR, Johnson S, Ghanem KG, Hook EW. The efficacy and safety of gentamicin plus azithromycin and gemifloxacin plus azithromycin as treatment of uncomplicated gonorrhea. Clin Infect Dis. 2014;59(8):1083–1091. doi: 10.1093/cid/ciu521. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 231. Ison CA, Deal C, Unemo M. Current and future treatment options for gonorrhea. 2013;89(Suppl 4):iv52–iv56. doi: 10.1136/sextrans-2012-050913. [ DOI ] [ PubMed ] [ Google Scholar ]
- 232. Brown LB, Krysiak R, Kamanga G, Mapanje C, Kanyamula H, Banda B, Mhango C, Hoffman M, Kamwendo D, Hobbs M, Hosseinipour MC, Martinson F, Cohen MS, Hoffman IF. Neisseria gonorrhoeae antimicrobial susceptibility in Lilonwe, Malawi, 2007. Sex Transm Dis. 2010;37(3):169–172. doi: 10.1097/olq.0b013e3181bf575c. [ DOI ] [ PubMed ] [ Google Scholar ]
- 233. Golparian D, Fernandes P, Ohnishi M, Jensen JS, Unemo M. In vitro activity of the new fluotoketolide solithromycin (CEM-101) against a large collection of clinical Neisseria gonorrhoeae isolates and international reference strains, including those with high-level antimicrobial resistance: potential treatment option for gonorrhea? Antimicrob Agents Chemo. 2012;56(5):2739–2742. doi: 10.1128/aac.00036-12. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 234. Unemo M, Golparian D, Limnios A, Whiley D, Ohnishi M, Lahra MM, Tapsall JW. In vitro activity of ertapenem versus ceftriaxone against Neisseria gonorrhoeae isolates with highly diverse ceftriaxone MIC values and effects of ceftriaxone resistance determinants: ertapenem for treatment of gonorrhea? Antimicrob Agents Chemo. 2012;56(7):3603–3609. doi: 10.1128/aac.00326-12. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 235. Jacobsson S, Golparian D, Phan LT, Ohnishi M, Fredlund H, Or YS, Unemo M. In vitro activities of the novel bicyclolides modithromycin (EDP-420, EP-013420, S-013420) and EDP-322 against MDR clinical Neisseria gonorrhoeae isolates and international reference strains. J Antimicrobial Chem. 2014;70(1):173–177. doi: 10.1093/jac/dku344. [ DOI ] [ PubMed ] [ Google Scholar ]
- 236. Falagas M, Karageorgopoulos D, Dimopoulos G. Clinical significance of the pharmacokinetic and pharmacodynamics characteristics of tigercycline. Cur Drug Metab. 2009;10(1):13–21. doi: 10.2174/138920009787048356. [ DOI ] [ PubMed ] [ Google Scholar ]
- 237. Nix DE, Matthias KR. Should tigecycline be considered for urinary tract infection? A pharmacokinetic re-evaluation . J Antimicrobial Chem. 2010;65(6):1311–1312. doi: 10.1093/jac/dkq116. [ DOI ] [ PubMed ] [ Google Scholar ]
- 238. Biedenbach DJ, Turner LL, Jones RN, Farrell DJ. Activity of JNJ-Q2, a novel floroquinolone, tested against Neisseria gonorrhoeae, including ciprofloxacin-resistant strains. Diagn Microbiol Infect Dis. 2012;74(2):204–206. doi: 10.1016/j.diagmicrobio.2012.06.006. [ DOI ] [ PubMed ] [ Google Scholar ]
- 239. Hamasuna R, Yasuda M, Ishikawa K, Uehara S, Hayami H, Takahashi S, Matsumoto T, Yamamoto S, Minamitani S, Watanabe A, Iwata S, Kaku M, Kadota J, Sunakawa K, Sato J, Hanaki H, Tsukamoto T, Kiyota H, Egawa S, Tanaka K, Arakawa S, Fujisawa M, Kumon H, Kobayashi K, Matsubara A, Naito S, Kuroiwa K, Hirayama H, Narita H, Hosobe T. The second nationwide surveillance of the antimicrobial susceptibility of Neisseria gonorrhoeae from male urethritis in Japan, 2012-2013. J Infect Chemo. 2015;21(5):340–345. doi: 10.1016/j.jiac.2015.01.010. [ DOI ] [ PubMed ] [ Google Scholar ]
- 240. Kazamori D, Aoi H, Sugimoto K, Ueshima T, Amano H, Itoh K, Kuramoto Y, Yazaki A. In vitro activity of WQ-3810, a novel fluoroquionolone, against multidrug-resistant and fluoroquinone-resistant pathogens. Int J Antimicrob Agents. 2014;44(5):443–449. doi: 10.1016/j.ijantimicag.2014.07.017. [ DOI ] [ PubMed ] [ Google Scholar ]
- 241. Fujimoto K, Takemoto K, Hatano K, Nakai T, Terashita S, Matsumoto M, Eriguchi Y, Eguchi K, Shimizudani T, Sato K, Kanazawa K, Sunagawa M, Ueda Y. Novel carbapenem antibiotics for parenteral and oral applications: in vitro and in vivo activites of 2-aryl carbapenems and their pharmacokinetics in laboratory animals. Antimicrob Agents Chemo. 2012;57(2):697–707. doi: 10.1128/aac.01051-12. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 242. Mendes RE, Alley MR, Sader HS, Biedenbach DJ, Jones RN. Potency and spectrum of activity of AN3365, a novel boron-containing protein synthesis inhibitor, tested against clinical isolates of Enterobacteriaceae and nonfermentative gram-negative bacilli. Antimicrob Agents Chemo. 2013;57(6):2849–2857. doi: 10.1128/aac.00160-13. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 243. Lomovskaya O, Watkins W. Inhibition of efflux pumps as a novel approach to combat drug resistance in bacteria. J Mol Microbiol Biotechnol. 2001;3(2):225–236. [ PubMed ] [ Google Scholar ]
- 244. Li L-H, Yen M-Y, Ho C-C, Wu P, Wang C-C, Maurya PK, Chen P-S, Chen W, Hsieh W-Y, Chen H-W. Non-cytotoxic nanomaterials enhance antimicrobial activities of cefmetazole against multidrug-resistant Neisseria gonorrhoeae. PLoS One. 2013;8(5):e64794. doi: 10.1371/journal.pone.0064794. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 245. Bucki R, Leszczynska K, Namiot A, Sokolowski W. Cathelicidin LL-37: A Multitask Antimicrobial Peptide. Arch Immunol Ther Exp (Warsz) 2010;58(1):15–25. doi: 10.1007/s00005-009-0057-2. [ DOI ] [ PubMed ] [ Google Scholar ]
- 246. Zhou P, Barb A. Mechanism and Inhibition of LpxC: An Essential Zinc-Dependent Deacetylase of Bacterial Lipid A Synthesis. Curr Pharm Biotechnol. 2008;9(1):9–15. doi: 10.2174/138920108783497668. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 247. Escaich S, Prouvensier L, Saccomani M, Durant L, Oxoby M, Gerusz V, Moreau F, Vongsouthi V, Maher K, Morrissey I, Soulama-Mouze C. The MUT056399 Inhibitor of FabI Is a New Antistaphylococcal Compound. Antimicrob Agents Chemo. 2011;55(10):4692–4697. doi: 10.1128/aac.01248-10. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 248. Jeverica S, Golparian D, Hanzelka B, Fowlie AJ, Mati M, Unemo M. High in vitro activity of a novel dual bacterial topoisomerase inhibitor of the ATPase activities of GyrB and ParE (VT12-008911) against Neisseria gonorrhoeae isolates with various high-level antimicrobial resistance and multidrug resistance. J Antimicrobial Chem. 2014;69(7):1866–1872. doi: 10.1093/jac/dku073. [ DOI ] [ PubMed ] [ Google Scholar ]
- 249. Unemo M, Shafer WM. Future treatment of gonorrhea - novel emerging drugs are essential an in progress? Expert Opin Emerg Drugs. 2015;20:357–360. doi: 10.1517/14728214.2015.1039981. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 250. Kern G, Palmer T, Ehmann DE, Shapiro AB, Andrews B, Basarab GS, Doig P, Fan J, Gao N, Mills SD, Mueller J, Sriram S, Thresher J, Walkup GK. Inhibition of Neisseria gonorrhoeae Type II Topoisomerases by the Novel Spiropyrimidinetrione AZD0914. J Biol Chem. 2015;290(34):20984–20994. doi: 10.1074/jbc.m115.663534. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 251. Alm RA, Lahiri SD, Kutschke A, Otterson LG, McLaughlin RE, Whiteaker JD, Lewis LA, Su X, Huband MD, Gardner H, Mueller JP. Characterization of the Novel DNA Gyrase Inhibitor AZD0914: Low Resistance Potential and Lack of Cross-Resistance in Neisseria gonorrhoeae. Antimicrob Agents Chemo. 2014;59(3):1478–1486. doi: 10.1128/aac.04456-14. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- 252. Goire N, Lahra MM, Chen M, Donovan B, Fairley CK, Guy R, Kaldor J, Regan D, Ward J, Nissen MD, Sloots TP, Whiley DM. Future treatment of gonorrhea - novel emerging drugs are essential an in progress? Expert Opin Emerg Drugs. 2014;12(3):223–229. doi: 10.1038/nrmicro3217. [ DOI ] [ PMC free article ] [ PubMed ] [ Google Scholar ]
- View on publisher site
- PDF (650.6 KB)
- Collections
Similar articles
Cited by other articles, links to ncbi databases.
- Download .nbib .nbib
- Format: AMA APA MLA NLM
Add to Collections
Affiliations
- 2 St. Luke's University Hospital
- PMID: 32644329
- Bookshelf ID: NBK558903
Neisseria gonorrhoeae , an obligate human pathogen, is a sexually transmitted disease that causes consequential worldwide morbidity both in resource-abundant and resource-limited nations, and its diagnosis and treatment require costly expenditures annually. Like other sexually transmitted infections (STIs), gonorrhea disproportionately impacts young adult populations. An ancient disease with biblical references (Hebrew Bible; Leviticus 15:1-3), gonorrhea has many slang references, including “the clap,” which likely derived from the name of the ancient Parisian red-light district Les Clapiers.
Copyright © 2024, StatPearls Publishing LLC.
- Continuing Education Activity
- Introduction
- Epidemiology
- Pathophysiology
- Histopathology
- History and Physical
- Treatment / Management
- Differential Diagnosis
- Complications
- Consultations
- Deterrence and Patient Education
- Enhancing Healthcare Team Outcomes
- Review Questions
Publication types
- Study Guide
- Search Menu
- Sign in through your institution
- Advance articles
- Editor's Choice
- Supplement Archive
- Cover Archive
- IDSA Guidelines
- IDSA Journals
- The Journal of Infectious Diseases
- Open Forum Infectious Diseases
- Photo Quizzes
- State-of-the-Art Reviews
- Voices of ID
- Author Guidelines
- Open Access
- Why Publish
- IDSA Journals Calls for Papers
- Advertising and Corporate Services
- Advertising
- Journals Career Network
- Reprints and ePrints
- Sponsored Supplements
- Branded Books
- About Clinical Infectious Diseases
- About the Infectious Diseases Society of America
- About the HIV Medicine Association
- IDSA COI Policy
- Editorial Board
- Self-Archiving Policy
- For Reviewers
- For Press Offices
- Journals on Oxford Academic
- Books on Oxford Academic
Article Contents
Results and discussion, research priorities.
- < Previous
Management of Gonorrhea in Adolescents and Adults in the United States
- Article contents
- Figures & tables
- Supplementary Data
Sarah Kidd, Kimberly A. Workowski, Management of Gonorrhea in Adolescents and Adults in the United States, Clinical Infectious Diseases , Volume 61, Issue suppl_8, December 2015, Pages S785–S801, https://doi.org/10.1093/cid/civ731
- Permissions Icon Permissions
Gonorrhea is the second most commonly reported notifiable disease in the United States and is associated with serious health sequelae, including pelvic inflammatory disease, infertility, and ectopic pregnancy. Treatment for gonorrhea has been complicated by antimicrobial resistance. Neisseria gonorrhoeae has developed resistance to each of the antimicrobials that were previously recommended as first-line treatment regimens, and current treatment options are severely limited. This article summarizes the key questions and data that were discussed at the Sexually Transmitted Diseases (STD) Treatment Guidelines Expert Consultation meeting in April 2013, and the rationale for the 2015 Centers for Disease Control and Prevention STD treatment guidelines for gonococcal infections in adolescents and adults. Key issues addressed include whether to change the dosage of ceftriaxone and azithromycin used in the recommended dual treatment regimen, whether to continue to list dual treatment with cefixime and azithromycin as an alternative treatment regimen, and management of gonococcal infections in persons with severe cephalosporin allergy or suspected treatment failure.
Gonorrhea, a sexually transmitted infection, is the second most commonly reported notifiable disease in the United States [ 1 ], with a total of 333 004 new cases reported to the Centers of Disease Control and Prevention (CDC) in 2013 [ 2 ]. However, because many infections are never diagnosed or reported, the true burden of gonococcal infection is likely significantly higher. It is estimated that >800 000 new gonococcal infections occur in the United States each year [ 3 ]. Although the national gonorrhea rate has declined substantially from its peak in 1975 (464.1 cases per 100 000 population) and reached an all-time low in 2009 (98.1 cases per 100 000), the rate subsequently increased each year during 2010–2012, and plateaued at 106.1 cases per 100 000 population in 2013 [ 2 ]. High gonorrhea rates continue to be observed in certain demographic groups and geographic areas. In particular, adolescents aged 15–19 years (337.5 cases per 100 000 population), young adults aged 20–24 years (500.5 cases per 100 000 population), non-Hispanic blacks (426.6 cases per 100 000 population), and residents of the Southern United States (128.6 cases per 100 000 population) bear the highest burden of disease [ 2 ].
Timely and effective treatment is an essential component of gonorrhea control programs. It reduces transmission in the community by shortening the duration of infection and decreases the risk of serious health sequelae, including pelvic inflammatory disease, infertility, and ectopic pregnancy [ 4 ]. Unfortunately, treatment for gonorrhea has been complicated by antimicrobial resistance. Neisseria gonorrhoeae has progressively developed resistance to each of the antimicrobials that were previously recommended as first-line treatment regimens, and current treatment options are severely limited [ 5 ]. In 2013, the CDC hosted a meeting with expert consultants to review the most recent data on gonococcal antimicrobial susceptibility, review the most recent data on treatment effectiveness, and make recommendations for the management of gonorrhea in adolescents and adults in the United States. This paper summarizes the evidence discussed at the meeting and the rationale for the 2015 CDC Sexually Transmitted Diseases (STD) treatment guidelines for gonococcal infections in adolescents and adults.
The CDC hosted a STD Treatment Guidelines Expert Consultation meeting in April 2013, where >60 experts in the fields of STD, infectious disease, epidemiology, and medicine discussed the latest developments in STD clinical and preventive services. In preparation for the meeting, 9 key questions on the management of gonococcal infections were developed based on input solicited from partners at state and local health departments and expert consultants. To address the key questions, a PubMed search for relevant articles published since 2008 (previous treatment guidelines meeting) through 9 March 2013 was conducted using the search terms “(gonorrhea or gonorrhoeae or gonorrrhoea or gonococcus or gonococcal) AND (treatment or therapy or resistance or antibiotics or failure)” and was restricted to abstracts or articles written in English. References listed in the retrieved articles were also searched for other relevant articles and abstracts. Additional data on national or regional gonococcal susceptibility were obtained from reports published on the websites of internationally recognized public health agencies and directly from the US Gonococcal Isolate Surveillance Project (GISP). GISP is a sentinel surveillance system, located in 25–30 cities throughout the United States, that monitors gonococcal antimicrobial susceptibility among urethral isolates obtained from men with urethritis [ 6 ]. Relevant data from these articles and reports were reviewed, summarized, and presented to a group of expert consultants at the April 2013 meeting. Preliminary answers to the 9 key questions and proposed recommendations were drafted based on available evidence or, when data were insufficient, expert opinion. Below is a list of key questions discussed, a summary of the data available for each question, and an overview of the discussion and recommendations resulting from each question.
In July 2013, the results of a clinical trial evaluating 2 new dual treatment regimens for gonorrhea were presented at an international conference and subsequently published [ 7 , 8 ]. In September 2013, a subset of the expert consultants met to discuss the data from the trial, and amended the proposed recommendations based on the new evidence. These data and revised recommendations are included in the following discussion.
Key Question 1. Are There Any Data to Suggest That There Should Be a Change in the Recommended Dosage of Ceftriaxone or Azithromycin for the Treatment of Uncomplicated Urethral, Cervical, or Rectal Gonococcal Infections?
At the time of the 2013 guidelines meeting, the CDC recommendation for treatment of uncomplicated gonococcal infection of the cervix, urethra, and rectum was dual treatment with ceftriaxone 250 mg intramuscularly as a single dose and either azithromycin 1 g orally as a single dose or doxycycline 100 mg orally twice daily for 7 days; azithromycin was preferred over doxycycline as the second antimicrobial, owing to the high prevalence of tetracycline resistance among gonococcal isolates in the United States [ 2 , 9 ]. However, other countries have recommended higher doses of ceftriaxone and/or azithromycin than those recommended by the CDC [ 10–12 ].
Ceftriaxone Clinical Effectiveness Data
The CDC has traditionally used the criteria of ≥95% effectiveness, and a lower 95% confidence interval (CI) bound of ≥95% effectiveness, for recommended treatment regimens [ 13 , 14 ]. According to summed data from clinical trials published in the 1980s and early 1990s, the effectiveness of ceftriaxone 250 mg for uncomplicated urethral, cervical, and rectal gonococcal infections is 99.2% (95% CI, 98.8%–99.5%) (Table 1 ) [ 21 ]. There are no new clinical trial data on the efficacy of ceftriaxone 250 mg. A literature review identified 2 recent studies assessing the clinical effectiveness of ceftriaxone at higher doses: one evaluated ceftriaxone 500 mg and one evaluated ceftriaxone 1 g. The evaluation of ceftriaxone 500 mg reported a cure rate of 90% in 100 patients with urethral or cervical infection [ 16 ]. However, this study did not use standard bacteriologic criteria to confirm gonococcal infection and treatment failure, and these results should be interpreted with caution. The evaluation of ceftriaxone 1 g demonstrated a cure rate of 100% in 48 patients with urethral or cervical gonococcal infection [ 17 ].
Clinical Effectiveness Data for the Treatment of Uncomplicated Gonococcal Infections of the Urethra, Cervix, or Rectum
Abbreviations: CI, confidence interval; IM, intramuscularly; IV, intravenously; MIC, minimum inhibitory concentration; PO, orally.
a Failure defined as persistence of symptoms with presence of gram-negative diplococci and pus on day 5 following treatment; no comment on evaluation for reexposure.
Ceftriaxone Susceptibility Data
In general, for an antimicrobial to meet the ≥95% clinical effectiveness criterion, ≥95% of gonococcal infections must be susceptible to the antimicrobial. Accordingly, although the correlation between in vitro resistance and clinical treatment failure is imperfect, in the past the CDC has changed treatment guidelines when the prevalence of resistance to a recommended antimicrobial reached ≥5% in the population [ 14 ]. The in vitro minimum inhibitory concentration (MIC) breakpoints that correspond to cefixime and ceftriaxone resistance have not been defined, but the Clinical and Laboratory Standards Institute (CLSI) defines decreased susceptibility to ceftriaxone as an MIC ≥0.5 µg/mL [ 23 ]. The proportion of GISP isolates with decreased susceptibility to ceftriaxone has remained <0.1% (Table 2 ). While increases in cephalosporin MICs were observed worldwide during 2000–2010 [ 9 , 24–26 ], ceftriaxone MICs in the United States during 2011–2013 were similar to ceftriaxone MICs during 1987–2000, when the majority of the ceftriaxone clinical trials were conducted (Table 2 ).
Ceftriaxone Minimum Inhibitory Concentrations in the Gonococcal Isolate Surveillance Project, United States, 1987–2013
Source: Gonococcal Isolate Surveillance Project, unpublished data.
Abbreviations: MIC 50 , minimum concentration needed to inhibit 50% of isolates; MIC 90 , minimum concentration needed to inhibit 90% of isolates.
Worldwide, a total of 4 isolates with very high ceftriaxone MICs (MICs 1–4 µg/mL) have been identified (Table 3 ) [ 33 , 37 , 38 , 42 , 43 ]. The first of these was associated with a pharyngeal infection and ceftriaxone 1 g treatment failure. The other 3 were associated with urethral or rectal infections that were treated with antimicrobials other than ceftriaxone. No other isolates with ceftriaxone MICs in this range or higher have been reported.
Cefixime Treatment Failures, Ceftriaxone Treatment Failures, and Additional Reports of High-Level Cefixime or Ceftriaxone Resistance
Abbreviations: IM, intramuscularly; IV, intravenously; MIC, minimum inhibitory concentration; PO, orally.
a Pretreatment and posttreatment isolates from 2 of these cases had discordant antibiograms, suggesting that these 2 cases were reinfections, not treatment failures.
b One urethral infection originally treated with cefixime 400 mg and doxycycline also failed retreatment with cefixime 400 mg PO × 1.
c One rectal infection originally treated with cefixime 400 mg PO × 1 also failed retreatment with cefixime 800 mg PO × 1.
d This rectal infection, originally treated with cefixime 800 mg, also failed retreatment with cefixime 400 mg PO × 1 plus doxycycline 100 mg PO twice daily × 7 days.
Case Reports of Ceftriaxone Treatment Failures
There have been no reported cases of ceftriaxone treatment failure for urethral, cervical, or rectal infection, and no reported cases of ceftriaxone treatment failure at any anatomic site in the United States (Table 3 ). While ceftriaxone treatment failure for pharyngeal infection has been reported at MICs as low as 0.016 µg/mL [ 36 ], pharyngeal infection is known to be more difficult to eradicate, and treatment failure at this site does not necessarily indicate antimicrobial resistance [ 22 ].
Pharmacokinetic Estimates of Ceftriaxone Effectiveness
A pharmacodynamic modeling study published in 2010 predicted that treatment failures with ceftriaxone 250 mg would be likely at ceftriaxone MICs of 0.125–0.25 µg/mL [ 44 ]. However, clinical data on the ceftriaxone MIC breakpoint that is correlated with treatment failure for urethral, cervical, or rectal infection are lacking.
Rationale for Dual Treatment
Prior to the 2010 STD treatment guidelines, administration of a second antimicrobial was recommended for patients with gonococcal infection to provide treatment for Chlamydia trachomatis infection, which frequently co-occurred with gonococcal infection [ 45 ]. Since the publication of the 2010 treatment guidelines, dual treatment that included either azithromycin or doxycycline has been recommended for all patients with gonococcal infection, regardless of the presence of chlamydial coinfection [ 46 ]. The primary rationale for recommending dual treatment is that it may enhance treatment effectiveness and prevent further transmission of a resistant organism. In addition, coadministration of 2 antimicrobials with different mechanisms of action may also hinder the development of resistance.
Recent recommendations have stated that azithromycin was preferred over doxycycline as the second antimicrobial owing to the high prevalence of tetracycline resistance among gonococcal isolates in the United States [ 9 , 47 ]. In 2013, 23.7% of GISP isolates were resistant to tetracycline [ 2 ].
Azithromycin Clinical Effectiveness Data
Based on summed data from clinical trials, monotherapy with azithromycin 1 g cures 97.6% of uncomplicated gonococcal infections of the urethra, cervix, or rectum (95% CI, 95.7%–98.9%), and monotherapy with azithromycin 2 g cures 99.2% of these infections (95% CI, 97.3%–99.9%) (Table 1 ) [ 14 ]. For urethral and cervical infections, a more recent review estimated that the clinical effectiveness of azithromycin 1 g was 96.5% (95% CI, 94.3%–97.6%) if retrospective studies were excluded and 97.0% (95% CI, 95.2%–97.9%) if retrospective studies were included [ 15 ]. The same review estimated that azithromycin 2 g cured 99.0% (95% CI, 97.5%–99.6%) of urethral or cervical infections.
Azithromycin Susceptibility Data
Azithromycin MICs among US GISP isolates appear to have slightly increased from the early 1990s, when the majority of azithromycin clinical studies were conducted, to 2011–2013 (Table 4 ). However, interpretation of these data is complicated by the use of a new media formulation beginning in 2005, which may have resulted in a one-dilution increase in azithromycin MIC results [ 48 ].
Azithromycin Minimum Inhibitory Concentrations in the Gonococcal Isolate Surveillance Project, United States, 1992–2013
In 2005, the Gonococcal Isolate Surveillance Project began using a new media formulation for azithromycin susceptibility testing, which resulted in a one-dilution increase in azithromycin MICs.
The MIC breakpoint that defines resistance or that correlates with clinical treatment failure has not been defined for azithromycin, and CLSI has not established an MIC breakpoint that defines decreased susceptibility to azithromycin. In GISP, isolates with azithromycin MICs ≥2 µg/mL are considered to have elevated MICs [ 6 ]. The percentage of GISP isolates with elevated azithromycin MICs has remained <1% (Table 4 ). However, of concern, sporadic cases of high-level azithromycin resistance (MICs ≥256 µg/mL) have been identified in the United Kingdom, Italy, Argentina, Hong Kong, and the United States [ 49–54 ].
Case Reports of Azithromycin Treatment Failures
There have been multiple reports of azithromycin treatment failures since the 1990s, but these are infrequently associated with higher azithromycin MICs and so do not necessarily indicate failure due to antimicrobial resistance [ 32 , 55–60 ]. Urethral and cervical infection treatment failures have been associated with pretreatment azithromycin MICs of 0.06–0.5 µg/mL following treatment with azithromycin 1 g and MICs of 0.25–1 µg/mL following treatment with azithromycin 2 g.
Other Considerations for Azithromycin Dosing
Other considerations for azithromycin include the ease with which N. gonorrhoeae develops resistance to macrolides when given as monotherapy. Most recently, a case report documented the rapid emergence of resistance (MIC increased from 1 µg/mL to 8 µg/mL) following a single 2-g dose of azithromycin [ 60 ].
In addition, when considering increasing the dual treatment dose of azithromycin from 1 g to 2 g as part of a dual treatment regimen, a tradeoff exists between the possible benefit of increasing the cure rate and the risk of increasing the frequency or severity of adverse effects. Depending on formulation, studies using azithromycin 2 g as a single dose report vomiting in 0.7%–7.0% of patients and gastrointestinal symptoms in up to 24.4%–35.3% [ 61–63 ]. In comparison, azithromycin 1 g as a single dose is generally associated with fewer and milder gastrointestinal symptoms, and studies using a 1-g dose report any adverse effect in <10% of patients [ 55 , 56 , 59 ].
Recommendations
The available clinical data indicate that ceftriaxone 250 mg is effective in approximately 99% of uncomplicated urethral, cervical, and rectal gonococcal infections. There are no clinical data to support the administration of ceftriaxone at higher doses than 250 mg. Therefore, dual treatment for gonorrhea that includes ceftriaxone at the 250-mg dose is recommended for the treatment of uncomplicated urethral, cervical, and rectal gonococcal infections.
When azithromycin 1 g is given as part of a dual treatment regimen with ceftriaxone, development and subsequent transmission of azithromycin resistance is unlikely. Therefore, based on the effectiveness of azithromycin 1 g and the increased adverse effects associated with the 2 g dose, azithromycin 1 g should be used when given as part of a dual treatment regimen with ceftriaxone.
Last, given the prevalence of tetracycline resistance among US GISP isolates, doxycycline is no longer recommended for use as the second antimicrobial for treatment of gonococcal infections. The recommended regimen for uncomplicated urethral, cervical, or rectal gonococcal infection is dual treatment with ceftriaxone 250 mg intramuscularly as a single dose and azithromycin 1 g orally as a single dose.
Key Question 2. Should Cefixime or Any Oral Cephalosporin Continue to Be Recommended as an Alternative Treatment for Urethral, Cervical, or Rectal Gonococcal Infections?
In 2012, CDC treatment guidelines were updated so that cefixime was no longer recommended as first-line treatment for gonorrhea [ 9 ]. This change was made based on observations that the overall percentage of GISP isolates with elevated cefixime MICs (MIC ≥0.25 µg/mL) had increased from 0.1% in 2006 to 1.5% during January–August 2011. Of particular concern was that the percentage of isolates with elevated cefixime MICs in the West increased from 0.2% to 3.2%, and the percentage among gay, bisexual, and other men who have sex with men (collectively referred to as MSM) in the West increased from 0.1% to 4.5%. Although the cefixime MIC breakpoints that correlate with clinical treatment failure have not been defined, there was concern that this pattern indicated early stages of the development of clinically significant gonococcal resistance to cefixime, and the rising cefixime MICs would soon result in declining effectiveness of cefixime. Additionally, there was concern that, as cefixime became less effective, its use might hasten the development of resistance to ceftriaxone. Although cefixime was no longer included as part of the recommended treatment regimen, CDC continued to list dual treatment with cefixime 400 mg orally as a single dose and either azithromycin 1 g orally as a single dose or doxycycline 100 mg orally twice daily for 7 days as an alternative regimen for gonococcal infections of the urethra, cervix, or rectum when ceftriaxone is not available.
Cefixime Clinical Effectiveness Data
Based on summed data from clinical trials published in the 1980s and 1990s, the effectiveness of cefixime 400 mg for urethral, cervical, and rectal gonococcal infections is 97.5% (95% CI, 95.4%–98.8%) [ 14 ]. The only recent data on cefixime effectiveness come from a retrospective analysis of gonococcal infections at any anatomic site (urethral, cervical, rectal, or pharyngeal) that were treated with a variety of cefixime-based regimens (cefixime 400 mg or 800 mg; some patients also received either azithromycin or doxycycline) (Table 1 ) [ 20 ]. Overall, in this analysis cefixime-based regimens cured 93.2% of gonococcal infections among patients who returned for test of cure, but cefixime effectiveness varied depending on cefixime MIC. Among patients who returned for test of cure, cefixime-based regimens successfully cured 98.1% of infections associated with a cefixime MIC <0.12 µg/mL, but only 75.0% of infections associated with a cefixime MIC ≥0.12 µg/mL (relative risk of treatment failure, 13.1 [95% CI, 2.9–59.7]). The authors also performed a secondary analysis to account for possible bias resulting from limiting the analysis to patients who returned for test of cure. If all patients who were treated were included in the analysis and it was assumed that no treatment failures occurred among those who did not return for tests of cure, cefixime-based regimens cured 99.1% of infections associated with a cefixime MIC <0.12 µg/mL, and 88.1% of infections associated with a cefixime MIC ≥0.12 µg/mL (relative risk of treatment failure 13.8 [95% CI, 2.9–64.5]).
Cefixime Susceptibility Data
The MIC breakpoint that corresponds to cefixime resistance and treatment failure has not been defined, but CLSI defines decreased susceptibility to cefixime as a cefixime MIC ≥0.5 µg/mL [ 23 ]. The proportion of US GISP isolates with decreased susceptibility to cefixime (MIC ≥0.5 µg/mL) has remained ≤0.1% (Table 5 ). In contrast, the proportion of GISP isolates with elevated cefixime MICs (MIC ≥0.25 µg/mL) has increased from 0.1% during 2001–2005 to 0.9% during 2011–2013. The proportion of isolates with MICs ≥0.125 µg/mL, the MIC threshold associated with increased risk of treatment failure [ 20 ], increased from 0.7% during 2001–2005 to 2.7% in 2011–2013.
Cefixime Minimum Inhibitory Concentrations in the Gonococcal Isolate Surveillance Project, United States, 1992–2013
a Cefixime susceptibility testing was not conducted during 2007–2008.
Case Reports of Cefixime Treatment Failures
Globally, cefixime treatment failures following treatment with cefixime 400 mg have generally been associated with cefixime MICs of 0.12–4 µg/mL [ 20 , 30–34 ] (Table 3 ). However, at least one study has reported cefixime treatment failures at MICs as low as ≤0.03 µg/mL [ 20 ].
Recommendation
Clinical data from a recent retrospective analysis and from documented cefixime treatment failures suggest that gonococcal infections with cefixime MICs ≥0.125 µg/mL are associated with a higher risk of treatment failure compared to those with MICs <0.125 µg/mL. Given the increase in cefixime MICs observed in the last decade, ceftriaxone is clearly preferable to cefixime for the treatment of gonococcal infections. However, there are no data to suggest that the clinical effectiveness of dual treatment with cefixime and azithromycin for urethral, cervical, and rectal gonococcal infections is <95% in the United States. Recognizing that there are circumstances where ceftriaxone is not available or where an injection is not possible, and that treatment with a cefixime-based dual treatment regimen is preferable to no treatment, dual treatment with cefixime 400 mg orally as a single dose and azithromycin 1 g orally as a single dose will continue to be an alternative regimen for the treatment of uncomplicated urethral, cervical, and rectal gonococcal infections when ceftriaxone is not available.
Key Question 3. Should Dual Treatment With Cefixime and Azithromycin Be Recommended for Expedited Partner Therapy (EPT)? Are There Any Data to Support Use of Azithromycin 2 g Over Azithromycin 1 g in Combination With Cefixime for EPT?
The 2012 update to CDC gonorrhea treatment guidelines recommended that EPT be considered for heterosexual partners of a patient with gonorrhea if they cannot be linked to evaluation and treatment in a timely fashion. In this scenario, EPT using dual treatment with cefixime 400 mg orally as a single dose and azithromycin 1 g orally as a single dose would be delivered to the partner by the patient, a disease investigation specialist, or through a collaborating pharmacy. The legal status of EPT varies by state. EPT has been shown to reduce the rate of reinfection among index patients and increase rates of partner treatment in clinical trials [ 64 ]. However, EPT requires an oral regimen, and given recent concerns about the continued effectiveness of cefixime, some have questioned whether the advantages of EPT outweigh the potential increased risk of treatment failures associated with a cefixime-based dual treatment regimen.
In practice, EPT must be prescribed without knowing the partner's complete history of sexual exposure, and there are no population data or estimates of the proportion of partners that are infected at different anatomic sites. It is therefore important to consider the effectiveness of an EPT regimen for urethral, cervical, and rectal infections, as well as pharyngeal infections. The previous question addressed the data on cefixime effectiveness for urethral, cervical, and rectal infections and concluded that dual treatment with cefixime and azithromycin should continue to be listed as an alternative treatment regimen in situations when ceftriaxone is not available (see Key Question 2). For pharyngeal infections, older summed clinical trials data estimate that monotherapy with cefixime 400 mg cures 92.3% (95% CI, 74.9%–95.7%), while ceftriaxone 250 mg cures 99.0% (95% CI, 94.4%–100%) of gonococcal infections of the pharynx (Table 6 ) [ 21 ]. More recent data on the effectiveness of cefixime for pharyngeal infections, and all data on the effectiveness of dual treatment regimens for pharyngeal infections, are limited to retrospective studies among the relatively small subset of persons who returned for test of cure. However, some retrospective data suggest that the effectiveness of dual treatment with an oral cephalosporin (either cefixime 400 mg or cefpodoxime 400 mg) in combination with azithromycin 1 g (93.0%) is comparable to the effectiveness of ceftriaxone 125–250 mg monotherapy (90.9%) or dual treatment with ceftriaxone 125–250 mg and either azithromycin 1 g or doxycycline (88.7%) [ 65 ].
Clinical Effectiveness Data for the Treatment of Uncomplicated Gonococcal Infections of the Pharynx
Data on the comparative effectiveness of azithromycin 1 g vs azithromycin 2 g as monotherapy, as well as their potential adverse effects, are reviewed elsewhere (see Key Question 1 for urethral, cervical, and rectal infections, and Key Question 7 for pharyngeal infections). There are no data comparing the effectiveness of dual treatment with cefixime and azithromycin 1 g vs dual treatment with cefixime and azithromycin 2 g for gonococcal infections at any anatomic site (Tables 1 and 6 ).
EPT increases rates of partner treatment and reduces reinfections among index patients. Although there are concerns about the effectiveness of cefixime-based regimens for the potential treatment of pharyngeal infection when used as EPT, these are outweighed by concerns that failure to use EPT will result in fewer partners receiving treatment. Based on these considerations, as well as the data presented in Key Question 2 that support continued use of dual treatment with cefixime and azithromycin as an alternative regimen for urethral, cervical, and rectal infections in settings where ceftriaxone is not available, dual treatment with cefixime 400 mg orally as a single dose and azithromycin 1 g orally as a single dose will continue to be recommended for EPT.
Key Question 4. Are There Any Other Drugs or Drug Regimens Besides Those Listed in the 2012 Update to the Treatment Guidelines That Can Be Recommended as First-line or Alternative Treatment for Gonorrhea?
At the time of the guidelines meeting, a literature search of studies published since 2008 did not identify any clinical trials for novel gonorrhea treatment regimens (Table 1 ). A systematic review of previously published clinical trials of gentamicin calculated that intramuscular gentamicin 240 mg or 280 mg had a pooled cure rate of 91.5% (95% CI, 88.1%–94.0%) [ 18 ] for urethral or cervical infection, lower than the CDC clinical effectiveness criterion. In addition, 2 studies assessed the clinical effectiveness of intramuscular spectinomycin 2 g for urogenital infection, and reported cure rates of 94% and 96.7% [ 16 , 19 ]. These data are consistent with older summed clinical trials data which estimate that the clinical effectiveness of spectinomycin is 98.2% (95% CI, 97.6%–99.9%) for urethral, cervical, and rectal gonococcal infections [ 21 ]. However, spectinomycin has poor efficacy against pharyngeal infection (51.8% [95% CI, 38.7%–64.9%]) (Table 6 ) [ 21 ], and is not currently available in the United States.
Following the guidelines meeting, the results of a clinical trial evaluating 2 new dual treatment regimens (dual treatment with gemifloxacin 320 mg orally as a single dose and azithromycin 2 g orally as a single dose, or dual treatment with gentamicin 240 mg intramuscularly as a single dose and azithromycin 2 g orally as a single dose) were published [ 8 ]. For urethral and cervical infections, this trial demonstrated that dual treatment with gemifloxacin and azithromycin cured 99.5% of infections (lower 95% CI bound, 97.6%) and dual treatment with gentamicin and azithromycin cured 100% of infections (lower 95% CI, bound 98.5%). Dual treatment with gemifloxacin and azithromycin also cured 15 of 15 pharyngeal and 5 of 5 rectal infections; dual treatment with gentamicin and azithromycin cured 10 of 10 pharyngeal and 1 of 1 rectal infections. Gastrointestinal adverse events were common with both regimens. Overall, 7.7% of patients given dual treatment with gemifloxacin and azithromycin and 3.3% of patients given dual treatment with gentamicin and azithromycin vomited within 1 hour of medication administration, necessitating retreatment with a different regimen.
Since 2008, there have been many published studies of in vitro activity of various agents against N. gonorrhoeae . Few agents have showed enough promise to warrant clinical trials. Notable agents tested in vitro include solithromycin [ 69 , 70 ], delafloxacin and other novel quinolones [ 71–75 ], ertapenem [ 76 ], and novel carbapenems [ 77 ]. Of these, only solithromycin and delafloxacin have progressed to phase 3 clinical trials (ClincalTrials.gov identifiers NCT02210325 and NCT02015637, respectively).
Based on data that became available after the treatment guidelines meeting, 2 new dual treatment regimens (dual treatment with gemifloxacin 320 mg orally as a single dose and azithromycin 2 g orally as a single dose, or dual treatment with gentamicin 240 mg intramuscularly as a single dose and azithromycin 2 g orally as a single dose) may be considered as alternative treatment options, but gastrointestinal adverse events may limit their use. Because of limited data on the effectiveness of these regimens for rectal and pharyngeal infections, and because of the frequency of gastrointestinal adverse events associated with these regimens, dual treatment with cefixime 400 mg orally as a single dose and azithromycin 1 g orally as a single dose is the preferred alternative regimen for urethral, cervical, and rectal infections if ceftriaxone is not available and the patient is not allergic to cephalosporins.
Key Question 5. What Regimen Should Be Recommended for Persons Who Fail Treatment With the Recommended Regimen (Dual Treatment With Ceftriaxone 250 mg and Azithromycin 1 g)?
Few ceftriaxone treatment failures have been identified worldwide (Table 3 ). Therefore, only minimal clinical experience is available to guide treatment recommendations for treatment failures following the recommended regimen. All documented ceftriaxone treatment failures have been pharyngeal infections, and have successfully resolved with either repeated or higher doses of ceftriaxone, dual treatment with ceftriaxone 250 mg and azithromycin 1 g, or single-dose azithromycin 2 g [ 36–41 ]. Urethral or rectal infections associated with high ceftriaxone MICs (1–2 µg/mL) resolved after treatment with either gentamicin, a course of levofloxacin followed by a multiday course of azithromycin, or a course of doxycycline.
When available, antimicrobial susceptibility results may guide treatment decisions. Other regimens recently demonstrated to have high clinical effectiveness, such as dual treatment with gemifloxacin and azithromycin or dual treatment with gentamicin and azithromycin (see Key Question 4) [ 8 ], may be of use in the management of ceftriaxone treatment failures.
Because the majority of suspected treatment failures are actually reinfections, persons with suspected treatment failure following treatment with the recommended regimen (dual treatment with ceftriaxone 250 mg intramuscularly as a single dose and azithromycin 1 g orally as a single dose) should usually be re-treated with the same regimen. However, in situations with a higher likelihood of treatment failure than reinfection, clinicians should (1) culture relevant clinical specimens and obtain antimicrobial susceptibility testing of any N. gonorrhoeae isolates, and advise the laboratory to retain the isolate(s) for possible further testing; (2) consult an infectious disease specialist, an STD/HIV Prevention Training Center clinical expert, the local or state health department, or CDC for advice on obtaining cultures, obtaining antimicrobial susceptibility testing, and treatment; and (3) report the case to CDC via the state or local health department. Clinicians may consider treating these patients with either dual treatment with gemifloxacin 320 mg orally as a single dose and azithromycin 2 g orally as a single dose, or dual treatment with gentamicin 250 mg intramuscularly as a single dose and azithromycin 2 g orally as a single dose. Clinicians should also obtain a test of cure at the relevant anatomic site 7–14 days after retreatment. Culture is the recommended test for test of cure, preferably with simultaneous nucleic acid amplification test (NAAT). Antimicrobial susceptibility testing should be performed if N. gonorrhoeae is isolated. All sex partners from the preceding 60 days should be evaluated promptly with culture and presumptively treated with the same regimen as the patient.
Key Question 6. What Regimens Should Be Recommended for the Treatment of Uncomplicated Urethral, Cervical, or Rectal Infection in Persons With Severe Cephalosporin Allergy?
At the time of the treatment guidelines meeting, the only available treatment option for patients with severe cephalosporin allergy that met CDC clinical effectiveness criteria was monotherapy with azithromycin 2 g [ 9 , 46 ]. Following the treatment guidelines meeting, the results of a clinical trial demonstrating the effectiveness of dual treatment with gemifloxacin 320 mg orally as a single dose and azithromycin 2 g orally as a single dose or dual treatment with gentamicin 240 mg intramuscularly as a single dose and azithromycin 2 g orally as a single dose were published (see Key Question 4) [ 8 ].
Given new data on the clinical effectiveness of 2 new dual treatment regimens (see Key Question 4) and the theoretical benefit of dual treatment using antimicrobials with different mechanisms of action (see Key Question 1), azithromycin 2 g is no longer recommended as an alternative regimen for persons with severe cephalosporin allergy. Dual treatment with gemifloxacin 320 mg orally as a single dose and azithromycin 2 g orally as a single dose, or dual treatment with gentamicin 240 mg intramuscularly as a single dose and azithromycin 2 g orally as a single dose are potential therapeutic options for these patients.
Key Question 7. Are Current Recommendations Sufficient for Pharyngeal Gonococcal Infection?
At the time of the guidelines meeting, the current (2012) recommendation for uncomplicated gonococcal infections of the pharynx was dual treatment with ceftriaxone 250 mg intramuscularly as a single dose and either azithromycin 1 g orally as a single dose or doxycycline 100 mg orally twice daily for 7 days [ 9 ]. Azithromycin was preferred over doxycycline as the second antimicrobial because of the high prevalence of tetracycline resistance among GISP isolates. There were no alternative regimens listed for treatment of pharyngeal infection.
Clinical Effectiveness Data
Gonococcal infections of the pharynx are more difficult to eradicate than infections of the urethra, cervix, or rectum [ 29 ], and few antimicrobial regimens reliably cure >90% of gonococcal pharyngeal infections (Table 6 ). According to summed data from clinical trials published in the 1980s and 1990s, ceftriaxone 250 mg eradicates 99.0% (95% CI, 94.4%–100%) of pharyngeal infections [ 21 ]. Prospective clinical trials data on azithromycin effectiveness for pharyngeal infections are particularly sparse. Older summed clinical trials data have estimated that the clinical effectiveness of azithromycin 1 g is 100% (95% CI, 29.2%–100%) and the effectiveness of azithromycin 2 g is 100% (95% CI, 82.3%–100%), but these estimates were based on only 3 and 19 infections, respectively [ 13 , 21 ]. A more recent systematic review found that azithromycin 1 g cured 100% (7/7) and azithromycin 2 g cured 97.5% (39/40) of pharyngeal infections [ 15 ].
Review of the literature published since 2008 identified just 2 new prospective clinical trials with data on pharyngeal infections. A clinical trial of ceftriaxone 1 g demonstrated 100% effectiveness in 25 of 25 patients [ 17 ]. In a second trial, dual treatment with gemifloxacin 320 mg and azithromycin 2 g cured 100% (15/15) of pharyngeal infections and dual treatment with gentamicin 240 mg and azithromycin 2 g cured 100% (10/10) pharyngeal infections [ 8 ]. Unfortunately, gastrointestinal adverse events were commonly associated with both dual treatment regimens, potentially limiting their routine use (see Key Question 4).
Several retrospective analyses have attempted to describe the effectiveness of various regimens for the treatment of gonococcal infections of the pharynx [ 65 , 67 , 68 ] (Table 6 ). Overall, dual treatment regimens that include either ceftriaxone or cefixime and azithromycin compare favorably to other regimens evaluated in these analyses. However, interpretation of these data is complicated by limitations associated with retrospective studies; data were restricted to the subset of patients who returned for test of cure, and there were relatively small numbers of infections evaluated for some treatment regimens. In addition, most of these analyses did not report whether patients had been reexposed between treatment and test of cure, so it is possible some apparent treatment failures in these studies were actually reinfections.
Case Reports of Ceftriaxone Treatment Failure
Pharyngeal infection treatment failures following treatment with ceftriaxone 250–500 mg monotherapy have been reported from Australia [ 36 , 40 ], Sweden [ 39 ], and Slovenia [ 41 ], and were associated with ceftriaxone MICs 0.03–0.25 µg/mL (Table 3 ). In addition, a pharyngeal infection treatment failure following treatment with ceftriaxone 1 g monotherapy was documented in Japan in 2009 and was associated with ceftriaxone MICs of 2–4 µg/mL [ 37 , 38 ].
There are insufficient data to suggest that any treatment regimen for gonococcal infections of the pharynx is more effective than dual treatment with ceftriaxone 250 mg and azithromycin 1 g. There are no data comparing the effectiveness of azithromycin 1 g vs 2 g when used in combination with ceftriaxone for treatment of pharyngeal infections. Although newer dual treatment regimens (ie, dual treatment with gemifloxacin and azithromycin or dual treatment with gentamicin and azithromycin) appear promising, there are insufficient data on the effectiveness of these regimens for pharyngeal infections, and the adverse effects associated with these regimens may limit their use in practice. Therefore, dual treatment with ceftriaxone 250 mg intramuscularly as a single dose and azithromycin 1 g orally as a single dose is the recommended regimen for uncomplicated infections of the pharynx.
Key Question 8. Are Current Treatment Recommendations Sufficient for Disseminated Gonococcal Infection?
At the time of the guidelines meeting, the recommended treatment regimen for disseminated gonococcal infection (DGI) without evidence of meningitis or endocarditis was ceftriaxone 1 g given intramuscularly or intravenously every 24 hours [ 46 ]. This regimen was to be continued for 24–48 hours after clinical improvement begins, at which time therapy could be switched to oral cefixime 400 mg twice daily to complete at least 1 week of antimicrobial therapy.
There are no recent studies published on the treatment of DGI. No treatment failures have been reported following treatment with the above regimens.
Given the absence of data on the topic, DGI recommendations are based on expert opinion. Owing to growing concerns over gonococcal antimicrobial resistance, all persons with suspected DGI should have relevant clinical specimens collected for gonococcal culture, and if positive, gonococcal antimicrobial susceptibility testing. Treatment for DGI should be guided by the results of antimicrobial susceptibility testing. Pending antimicrobial susceptibility results, treatment decisions should be made on the basis of clinical presentation. The duration of treatment for DGI has not been systematically studied and should be determined in consultation with an infectious disease specialist.
For DGI without meningitis or endocarditis, the recommended treatment is ceftriaxone 1 g intramuscularly or intravenously every 24 hours plus azithromycin 1 g orally as a single dose. Clinicians may consider switching to an oral agent 24–48 hours after substantial improvement, but choice of oral antimicrobial should be guided by the results of antimicrobial susceptibility testing. The total duration of antimicrobial treatment should be at least 7 days.
For gonococcal meningitis or endocarditis, the recommended treatment is ceftriaxone 1–2 g intravenously every 12–24 hours and azithromycin 1 g orally in a single dose. Parenteral treatment for meningitis should be continued for 10–14 days. Parenteral treatment for endocarditis should be continued for at least 4 weeks.
Key Question 9. Should a Test of Cure Be Performed After Treatment for Gonococcal Infection? If so, Then What Test Should Be Used for Test of Cure and How Soon After Treatment Should Test of Cure Be Performed?
In the 2012 update to the gonorrhea treatment guidelines, test of cure was recommended for (1) persons treated with an alternative regimen and (2) persons with suspected treatment failure following treatment with the recommended regimen [ 9 ]. For persons treated with an alternative regimen, test of cure using culture or NAAT was recommended 1 week after completion of treatment. For patients with suspected treatment failure, culture and antimicrobial susceptibility testing was recommended to document persistent infection and guide therapy, and test of cure was recommended 1 week after completion of retreatment.
Use of Culture Versus NAAT for Test of Cure
The use of culture for test of cure facilitates antimicrobial susceptibility testing. However, the sensitivity of NAATs for detection of N. gonorrhoeae is superior to culture [ 78–84 ]. In practice, gonococcal culture is not readily available, and it is likely that NAATs will be used for test of cure in most settings.
Timing of Test of Cure
Test of cure using NAATs is complicated by the fact that residual nucleic acid from nonviable bacteria can be detected by NAATs for a period of time after successful treatment. There are few data on the duration of persistent N. gonorrhoeae nucleic acid after successful treatment. One study, using a ligase chain reaction (LCR) test that is no longer marketed in the United States, found the median time to a negative urine LCR test was 1 day for men and 2 days for women [ 85 ]. In this study, >90% of tests were negative on day 5 after treatment, but 18% of patients continued to have intermittent shedding of gonococcal nucleic acids after their first negative test. A second study, using an in-house porA pseudogene polymerase chain reaction test, asked persons with urogenital gonorrhea to return for test of cure 4–7 days after treatment and found that 84% (16/19) of those who returned during this interval had a negative test [ 86 ]. Of patients with positive tests of cure, 2 had a negative test when they returned on day 11, and one did not return until day 19, at which time his test was negative. The third and most recent study with data on test of cure evaluated the APTIMA Combo 2 Assay and evaluated MSM who returned for test of cure 3–21 days after treatment [ 87 ]. In this study, tests of cure were positive in no (0/95) urethral infections, 7.4% (10/135) of rectal infections, and 5.2% (7/134) of pharyngeal infections. All positive rectal tests of cure were among persons who were tested within 14 days of treatment; all positive pharyngeal tests of cure were among persons who were tested within 10 days of treatment. Together, these results suggest that residual DNA typically clears from the urogenital site within 7 days, but may persist for longer at extragenital sites.
Culture or NAAT can be used for test of cure. In practice, it is likely that NAATs will be used for test of cure in most settings. If a NAAT test of cure is positive, every effort should be made to obtain confirmatory culture before retreatment, and all positive test of cure cultures should undergo antimicrobial susceptibility testing.
Given the evidence that dual treatment with the alternative treatment regimen (cefixime 400 mg and azithromycin 1 g) is most likely ≥95% effective for treatment of urethral, cervical, or rectal infections (see Key Question 2) as well as concerns about a low positive predictive value of NAAT tests of cure, routine test of cure for persons diagnosed with urogenital or rectal gonorrhea who are treated with the alternative regimen is not recommended. However, because of concerns over cefixime's effectiveness at the pharyngeal site (see Key Question 3), test of cure is recommended for persons diagnosed with pharyngeal infection who are treated with the alternative regimen. Test of cure is also recommended for persons with suspected treatment failure at any anatomic site of infection.
Based on the limited data on appropriate timing of test of cure using NAATs, test of cure should be performed 14 days after treatment in the setting of pharyngeal infections treated with the alternative regimen and 7–14 days after retreatment in the setting of suspected treatment failure.
Further research is needed to inform future recommendations for the optimal management of gonorrhea. Research priorities identified at the 2013 meeting included (1) evaluation of novel oral antimicrobials or novel combinations of antimicrobials for treatment of gonorrhea; (2) pharmacokinetic models for ceftriaxone and azithromycin in the treatment of gonorrhea at urethral, cervical, rectal, and pharyngeal sites; and (3) evaluation of transport media for gonococcal culture, to facilitate access to gonococcal culture and antimicrobial susceptibility testing.
Dual treatment with ceftriaxone 250 mg intramuscularly as a single dose and azithromycin 1 g orally as a single dose is recommended for the treatment of uncomplicated gonorrhea of the urethra, cervix, rectum, or pharynx. For urethral, cervical, and rectal infections, dual treatment with cefixime 400 mg orally as a single dose and azithromycin 1 g orally as a single dose may be used as an alternative regimen when ceftriaxone is not available. Owing to the high prevalence of tetracycline resistance in the United States, doxycycline is no longer recommended as a second antimicrobial in either the first-line or alternative dual treatment regimen. Test of cure will continue to be recommended for persons with pharyngeal infection who receive an alternative treatment regimen, but is no longer recommended for persons with urethral, cervical, or rectal infection who are treated with the alternative regimen. Based on recent data demonstrating the effectiveness of 2 new dual treatment regimens (dual treatment with either gemifloxacin 320 mg orally as a single dose and azithromycin 2 g orally as a single dose, or dual treatment with gentamicin 240 mg intramuscularly as a single dose and azithromycin 2 g orally as a single dose), these regimens may be considered for persons with cephalosporin allergy or for those persons who fail treatment following the recommended regimen. Monotherapy with azithromycin 2 g orally as a single dose is no longer recommended for patients with cephalosporin allergy. Further research to identify new antimicrobials or new combinations of antimicrobials for the treatment of gonorrhea, particularly oral regimens, is urgently needed.
Acknowledgments. We thank Hunter Handsfield, Ned Hook, Bob Kirkcaldy, Fred Sparling, and Jonathan Zenilman for providing their input on the key questions and their assistance in the development of the treatment guidelines.
Supplement sponsorship. This article appears as part of the supplement “Evidence Papers for the CDC Sexually Transmitted Diseases Treatment Guidelines,” sponsored by the Centers for Disease Control and Prevention.
Potential conflict of interest. Both authors: No reported conflicts.
Both authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.
Adams DA , Jajosky RA , Ajani U et al. . Summary of notifiable diseases—United States, 2012 . MMWR Morb Mortal Wkly Rep 2014 ; 61 : 1 – 121 .
Google Scholar
Centers for Disease Control and Prevention . Sexually transmitted disease surveillance 2013 . Atlanta, GA : US Department of Health and Human Services , 2014 .
Satterwhite CL , Torrone E , Meites E et al. . Sexually transmitted infections among US women and men: prevalence and incidence estimates, 2008 . Sex Transm Dis 2013 ; 40 : 187 – 93 .
Google Preview
Hook EW , Handsfield HH . Gonococcal infections in the adult . In: Holmes KK , Sparling PF , Stamm WE et al. . eds. Sexually transmitted diseases . 4th ed. McGraw-Hill , 2008 : 627 – 45 .
Unemo M , Shafer W . Antibiotic resistance in Neisseria gonorrhoeae : origin, evolution, and lessons learned for the future . Ann N Y Acad Sci 2011 ; 1230 : E19 – 28 .
Centers for Disease Control and Prevention . Gonococcal Isolate Surveillance Project (GISP) protocol . Available at: http://www.cdc.gov/std/gisp/GISP-Protocol-May-2014.pdf . Accessed 5 September 2015 .
Kirkcaldy RD . Treatment of gonorrhoea in an era of emerging cephalosporin resistance and results of a randomised trial of new potential treatment options . In: STI & AIDS World Congress , Vienna, Austria , 2013 .
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 .
Centers for Disease Control and Prevention . Update to CDC's sexually transmitted diseases treatment guidelines, 2010: oral cephalosporins no longer a recommended treatment for gonococcal infections . MMWR Morb Mortal Wkly Rep 2012 ; 61 : 590 – 4 .
Bignell C , Fitzgerald M . UK national guideline for the management of gonorrhoea in adults, 2011 . Int J STD AIDS 2011 ; 22 : 541 – 7 .
Bignell C , Unemo M . 2012 European guideline on the diagnosis and treatment of gonorrhoea in adults . Int J STD AIDS 2013 ; 24 : 85 – 92 .
Tanaka M . Emergence of multidrug-resistant Neisseria gonorrhoeae strains circulating worldwide . Int J Urol 2012 ; 19 : 98 – 9 .
Moran J , Levine W . Drugs of choice for the treatment of uncomplicated gonococcal infections . Clin Infect Dis 1995 ; 20 ( suppl 1 ): S57 – 65 .
Newman LM , Moran JS , Workowski KA . Update on the management of gonorrhea in adults in the United States . Clin Infect Dis 2007 ; 44 ( suppl 3 ): S84 – 101 .
Bignell C , Garley J . Azithromycin in the treatment of infection with Neisseria gonorrhoeae . Sex Transm Infect 2010 ; 86 : 422 – 6 .
Shams-ur Rehman , Khan A , Amanullah , Akhter K . Clinical efficacy of the various drugs used in the treatment of gonorrhoeae . J Ayub Med Coll Abbottabad 2009 ; 21 : 28 – 30 .
Muratani T , Inatomi H , Ando Y , Kawai S , Akasaka S , Matsumoto T . Single dose 1g ceftriaxone for urogenital and pharyngeal infection caused by Neisseria gonorrhoeae . Int J Urol 2008 ; 15 : 837 – 42 .
Dowell D , Kirkcaldy RD . Effectiveness of gentamicin for gonorrhoea treatment: systematic review and meta-analysis . Sex Transm Infect 2012 ; 88 : 589 – 94 .
Kojima M , Masuda K , Yada Y , Hayase Y , Muratani T , Matsumoto T . Single-dose treatment of male patients with gonococcal urethritis using 2g spectinomycin: microbiological and clinical evaluations . Int J Antimicrob Agents 2008 ; 32 : 50 – 4 .
Allen VG , Mitterni L , Seah C et al. . Neisseria gonorrhoeae treatment failure and susceptibility to cefixime in Toronto, Canada . JAMA 2013 ; 309 : 163 – 70 .
Moran JS . Gonorrhoea . BMJ Clin Evid 2007 ; 2007 .
Moran JS . Treating uncomplicated Neisseria gonorrhoeae infections: is the anatomic site of infection important? Sex Transm Dis 1995 ; 22 : 39 – 47 .
Clinical and Laboratory Standards Institute . Performance standards for antimicrobial susceptibility testing; 24th informational supplement. CLSI document M100-S24 . Wayne, PA : CLSI , 2014 .
European Centre for Disease Prevention and Control . Gonococcal antimicrobial susceptibility surveillance in Europe, 2011 . Stockholm : ECDC , 2013 .
Lahra MM . Australian Gonococcal Surveillance programme annual report, 2012 . Commun Dis Intell 2013 ; 37 : E233 – 9 .
Ito M , Yasuda M , Yokoi S et al. . Remarkable increase in central Japan in 2001–2002 of Neisseria gonorrhoeae isolates with decreased susceptibility to penicillin, tetracycline, oral cephalosporins, and fluoroquinolones . Antimicrob Agents Chemother 2004 ; 48 : 3185 – 7 .
Deguchi T , Yasuda M , Yokoi S et al. . Treatment of uncomplicated gonococcal urethritis by double-dosing of 200mg cefixime at a 6-h interval . J Infect Chemother 2003 ; 9 : 35 – 9 .
Yokoi S , Deguchi T , Ozawa T et al. . Threat to cefixime treatment for gonorrhea . Emerg Infect Dis 2007 ; 13 : 1275 – 7 .
Ota KV , Fisman DN , Tamari IE et al. . Incidence and treatment outcomes of pharyngeal Neisseria gonorrhoeae and Chlamydia trachomatis infections in men who have sex with men: a 13-year retrospective cohort study . Clin Infect Dis 2009 ; 48 : 1237 – 43 .
Forsyth S , Penney P , Rooney G . Cefixime-resistant Neisseria gonorrhoeae in the UK: a time to reflect on practice and recommendations . Int J STD AIDS 2011 ; 22 : 296 – 7 .
Unemo M , Golparian D , Syversen G , Vestrheim DF , Moi H . Two cases of verified clinical failures using internationally recommended first-line cefixime for gonorrhoea treatment, Norway, 2010 . Euro Surveill 2010 ; 15 .
Ison CA , Hussey J , Sankar KN , Evans J , Alexander S . Gonorrhoea treatment failures to cefixime and azithromycin in England, 2010 . Euro Surveill 2011 ; 16 .
Unemo M , Golparian D , Nicholas R , Ohnishi M , Gallay A , Sednaoui P . 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 .
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 .
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 .
Tapsall J , Read P , Carmody C et al. . Two cases of failed ceftriaxone treatment in pharyngeal gonorrhoea verified by molecular microbiological methods . Int J Med Microbiol 2009 ; 58 ( pt 5 ): 683 – 7 .
Ohnishi M , Saika T , Hoshina S et al. . Ceftriaxone-resistant Neisseria gonorrhoeae , Japan . Emerg Infect Dis 2011 ; 17 : 148 – 9 .
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 .
Unemo M , Golparian D , Hestner A . Ceftriaxone treatment failure of pharyngeal gonorrhoea verified by international recommendations, Sweden, July 2010 . Euro Surveill 2011 ; 16 : pii:19792 .
Chen MY , Stevens K , Tideman R et al. . Failure of 500mg of ceftriaxone to eradicate pharyngeal gonorrhoea, Australia . J Antimicrob Chemother 2013 ; 68 : 1445 – 7 .
Unemo M , Golparian D , Potocnik M , Jeverica S . Treatment failure of pharyngeal gonorrhoea with internationally recommended first-line ceftriaxone verified in Slovenia, September 2011 . Euro Surveill 2012 ; 17 : pii:20200 .
Carnicer-Pont D , Smithson A , Fina-Homar E , Bastida MT . First cases of Neisseria gonorrhoeae resistant to ceftriaxone in Catalonia, Spain, May 2011 . Enferm Infecc Microbiol Clin 2012 ; 30 : 218 – 9 .
Camara J , Serra J , Ayats J et al. . Molecular characterization of two high-level ceftriaxone-resistant Neisseria gonorrhoeae isolates detected in Catalonia, Spain . J Antimicrob Chemother 2012 ; 67 : 1858 – 60 .
Chisholm SA , Mouton JW , Lewis DA , Nichols T , Ison CA , Livermore DM . Cephalosporin MIC creep among gonococci: time for a pharmacodynamic rethink? J Antimicrob Chemother 2010 ; 65 : 2141 – 8 .
Centers for Disease Control and Prevention . Sexually transmitted diseases treatment guidelines, 2006 . MMWR Morb Mortal Wkly Rep 2006 ; 55 ( RR-11 ): 42 – 8 .
Centers for Disease Control and Prevention . Sexually transmitted diseases treatment guidelines, 2010 . MMWR Morb Mortal Wkly Rep 2010 ; 59 ( RR-12 ): 49 – 55 .
Kirkcaldy RD , Kidd S , Weinstock HS , Papp JR , Bolan GA . Trends in antimicrobial resistance in Neisseria gonorrhoeae in the USA: the Gonococcal Isolate Surveillance Project (GISP), January 2006-June 2012 . Sex Transm Infect 2013 ; 89 ( suppl 4 ): iv5 – 10 .
Centers for Disease Control and Prevention . Sexually transmitted disease surveillance 2005 Supplement. Gonococcal Isolate Surveillance Project (GISP) Annual Report 2005 . Atlanta, GA : US Department of Health and Human Services , 2007 .
Palmer HM , Young H , Winter A , Dave J . Emergence and spread of azithromycin-resistant Neisseria gonorrhoeae in Scotland . J Antimicrob Chemother 2008 ; 62 : 490 – 4 .
Starnino S , Stefanelli P . Azithromycin-resistant Neisseria gonorrhoeae strains recently isolated in Italy . J Antimicrob Chemother 2009 ; 63 : 1200 – 4 .
Galarza PG , Alcala B , Salcedo C et al. . Emergence of high level azithromycin-resistant Neisseria gonorrhoeae strain isolated in Argentina . Sex Transm Dis 2009 ; 36 : 787 – 8 .
Chisholm SA , Neal TJ , Alawattegama AB , Birley HD , Howe RA , Ison CA . Emergence of high-level azithromycin resistance in Neisseria gonorrhoeae in England and Wales . J Antimicrob Chemother 2009 ; 64 : 353 – 8 .
Lo JY , Ho KM , Lo AC . Surveillance of gonococcal antimicrobial susceptibility resulting in early detection of emerging resistance . J Antimicrob Chemother 2012 ; 67 : 1422 – 6 .
Katz AR , Komeya AY , Soge OO et al. . Neisseria gonorrhoeae with high-level resistance to azithromycin: case report of the first isolate identified in the United States . Clin Infect Dis 2012 ; 54 : 841 – 3 .
Steingrimsson O , Olafsson JH , Thorarinsson H , Ryan RW , Johnson RB , Tilton RC . Azithromycin in the treatment of sexually transmitted disease . J Antimicrob Chemother 1990 ; 25 ( suppl A ): 109 – 14 .
Waugh MA . Open study of the safety and efficacy of a single oral dose of azithromycin for the treatment of uncomplicated gonorrhoea in men and women . J Antimicrob Chemother 1993 ; 31 ( suppl E ): 193 – 8 .
Young H , Moyes A , McMillan A . Azithromycin and erythromycin resistant Neisseria gonorrhoeae following treatment with azithromycin . Int J STD AIDS 1997 ; 8 : 299 – 302 .
Tapsall JW , Shultz TR , Limnios EA , Donovan B , Lum G , Mulhall BP . Failure of azithromycin therapy in gonorrhea and discorrelation with laboratory test parameters . Sex Transm Dis 1998 ; 25 : 505 – 8 .
Swanston WH , Prabhakar P , Barrow L , Mahabir BS , Furlonge C . Single dose (direct observed) azithromycin therapy for Neisseria gonorrhoeae and Chlamydia trachomatis in STD clinic attenders with genital discharge in Trinidad and Tobago . West Indian Med J 2001 ; 50 : 198 – 202 .
Soge OO , Harger D , Schafer S et al. . Emergence of increased azithromycin resistance during unsuccessful treatment of Neisseria gonorrhoeae infection with azithromycin (Portland, OR, 2011) . Sex Transm Dis 2012 ; 39 : 877 – 9 .
Handsfield HH , Dalu ZA , Martin DH , Douglas JM Jr , McCarty JM , Schlossberg D . Multicenter trial of single-dose azithromycin vs. ceftriaxone in the treatment of uncomplicated gonorrhea. Azithromycin Gonorrhea Study Group . Sex Transm Dis 1994 ; 21 : 107 – 11 .
Riedner G , Rusizoka M , Todd J et al. . Single-dose azithromycin versus penicillin G benzathine for the treatment of early syphilis . N Engl J Med 2005 ; 353 : 1236 – 44 .
Hook EW III , Behets F , Van Damme K et al. . A phase III equivalence trial of azithromycin versus benzathine penicillin for treatment of early syphilis . J Infect Dis 2010 ; 201 : 1729 – 35 .
Trelle S , Shang A , Nartey L , Cassell JA , Low N . Improved effectiveness of partner notification for patients with sexually transmitted infections: systematic review . BMJ 2007 ; 334 : 354 .
Barbee LA , Kerani RP , Dombrowski JC , Soge OO , Golden MR . A retrospective comparative study of 2-drug oral and intramuscular cephalosporin treatment regimens for pharyngeal gonorrhea . Clin Infect Dis 2013 ; 56 : 1539 – 45 .
Manavi K , Zafar F , Shahid H . Oropharyngeal gonorrhoea: rate of co-infection with sexually transmitted infection, antibiotic susceptibility and treatment outcome . Int J STD AIDS 2010 ; 21 : 138 – 40 .
Sathia L , Ellis B , Phillip S , Winston A , Smith A . Pharyngeal gonorrhoea—is dual therapy the way forward? Int J STD AIDS 2007 ; 18 : 647 – 8 .
McMillan A , Young H . The treatment of pharyngeal gonorrhoea with a single oral dose of cefixime . Int J STD AIDS 2007 ; 18 : 253 – 4 .
Golparian D , Fernandes P , Ohnishi M , Jensen JS , Unemo M . In vitro activity of the new fluoroketolide solithromycin (CEM-101) against a large collection of clinical Neisseria gonorrhoeae isolates and international reference strains, including those with high-level antimicrobial resistance: potential treatment option for gonorrhea? Antimicrob Agents Chemother 2012 ; 56 : 2739 – 42 .
Putnam SD , Castanheira M , Moet GJ , Farrell DJ , Jones RN . CEM-101, a novel fluoroketolide: antimicrobial activity against a diverse collection of gram-positive and gram-negative bacteria . Diagn Microbiol Infect Dis 2010 ; 66 : 393 – 401 .
Jones RN , Biedenbach DJ , Ambrose PG , Wikler MA . Zabofloxacin (DW-224a) activity against Neisseria gonorrhoeae including quinolone-resistant strains . Diagn Microbiol Infect Dis 2008 ; 62 : 110 – 2 .
Roberts MC , Remy JM , Longcor JD , Marra A , Sun E , Duffy EM . In vitro activity of delafloxacin against Neisseria gonorrhoeae clinical isolates . In: STI & AIDS World Congress . Vienna, Austria , 2013 .
Lauderdale TL , Shiau YR , Lai JF , Chen HC , King CH . Comparative in vitro activities of nemonoxacin (TG-873870), a novel nonfluorinated quinolone, and other quinolones against clinical isolates . Antimicrob Agents Chemother 2010 ; 54 : 1338 – 42 .
Biedenbach DJ , Turner LL , Jones RN , Farrell DJ . Activity of JNJ-Q2, a novel fluoroquinolone, tested against Neisseria gonorrhoeae , including ciprofloxacin-resistant strains . Diagn Microbiol Infect Dis 2012 ; 74 : 204 – 6 .
Jones RN , Fritsche TR , Sader HS . Antimicrobial activity of DC-159a, a new fluoroquinolone, against 1,149 recently collected clinical isolates . Antimicrob Agents Chemother 2008 ; 52 : 3763 – 75 .
Unemo M , Golparian D , Limnios A et al. . In vitro activity of ertapenem versus ceftriaxone against Neisseria gonorrhoeae isolates with highly diverse ceftriaxone MIC values and effects of ceftriaxone resistance determinants: ertapenem for treatment of gonorrhea? Antimicrob Agents Chemother 2012 ; 56 : 3603 – 9 .
Fujimoto K , Takemoto K , Hatano K et al. . Novel carbapenem antibiotics for parenteral and oral applications: in vitro and in vivo activities of 2-aryl carbapenems and their pharmacokinetics in laboratory animals . Antimicrob Agents Chemother 2013 ; 57 : 697 – 707 .
Centers for Disease Control and Prevention . Recommendations for the laboratory-based detection of Chlamydia trachomatis and Neisseria gonorrhoeae —2014 . MMWR Recomm Rep 2014 ; 63 ( RR-02 ): 1 – 19 .
Schachter J , Moncada J , Liska S , Shayevich C , Klausner JD . Nucleic acid amplification tests in the diagnosis of chlamydial and gonococcal infections of the oropharynx and rectum in men who have sex with men . Sex Transm Dis 2008 ; 35 : 637 – 42 .
Mimiaga MJ , Mayer KH , Reisner SL et al. . Asymptomatic gonorrhea and chlamydial infections detected by nucleic acid amplification tests among Boston area men who have sex with men . Sex Transm Dis 2008 ; 35 : 495 – 8 .
Bachmann LH , Johnson RE , Cheng H , Markowitz LE , Papp JR , Hook EW III . Nucleic acid amplification tests for diagnosis of Neisseria gonorrhoeae oropharyngeal infections . J Clin Microbiol 2009 ; 47 : 902 – 7 .
Bachmann LH , Johnson RE , Cheng H et al. . Nucleic acid amplification tests for diagnosis of Neisseria gonorrhoeae and Chlamydia trachomatis rectal infections . J Clin Microbiol 2010 ; 48 : 1827 – 32 .
Bissessor M , Tabrizi SN , Fairley CK et al. . Differing Neisseria gonorrhoeae bacterial loads in the pharynx and rectum in men who have sex with men: implications for gonococcal detection, transmission, and control . J Clin Microbiol 2011 ; 49 : 4304 – 6 .
Bromhead C , Miller A , Jones M , Whiley D . Comparison of the cobas 4800 CT/NG test with culture for detecting Neisseria gonorrhoeae in genital and nongenital specimens in a low-prevalence population in New Zealand . J Clin Microbiol 2013 ; 51 : 1505 – 9 .
Bachmann LH , Desmond RA , Stephens J et al. . Duration of persistence of gonococcal DNA detected by ligase chain reaction in men and women following recommended therapy for uncomplicated gonorrhea . J Clin Microbiol 2002 ; 40 : 3596 – 601 .
Hjelmevoll SO , Olsen ME , Sollid JU et al. . Appropriate time for test-of-cure when diagnosing gonorrhoea with a nucleic acid amplification test . Acta Derm Venereol 2012 ; 92 : 316 – 9 .
Beymer MR , Llata E , Stirland AM et al. . Evaluation of gonorrhea test of cure at 1 week in a Los Angeles community-based clinic serving men who have sex with men . Sex Transm Dis 2014 ; 41 : 595 – 600 .
- ceftriaxone
- azithromycin
- gonococcal infection
- centers for disease control and prevention (u.s.)
- treatment failure
- antimicrobials
- treatment guidelines
Email alerts
More on this topic, related articles in pubmed, citing articles via, looking for your next opportunity.
- X (formerly Twitter)
- Recommend to your Library
Affiliations
- Online ISSN 1537-6591
- Print ISSN 1058-4838
- Copyright © 2024 Infectious Diseases Society of America
- About Oxford Academic
- Publish journals with us
- University press partners
- What we publish
- New features
- Open access
- Institutional account management
- Rights and permissions
- Get help with access
- Accessibility
- Media enquiries
- Oxford University Press
- Oxford Languages
- University of Oxford
Oxford University Press is a department of the University of Oxford. It furthers the University's objective of excellence in research, scholarship, and education by publishing worldwide
- Copyright © 2024 Oxford University Press
- Cookie settings
- Cookie policy
- Privacy policy
- Legal notice
This Feature Is Available To Subscribers Only
Sign In or Create an Account
This PDF is available to Subscribers Only
For full access to this pdf, sign in to an existing account, or purchase an annual subscription.
IMAGES
VIDEO
COMMENTS
Gonorrhoea is a sexually transmitted infection (STI) caused by Neisseria gonorrhoeae (the gonococcus). In 2016, an estimated 87 million incident cases occurred among persons aged 15–49 years worldwide with an incidence rate of 20 cases/1000 women and 26/1000 men [1].
Neisseria gonorrhoeae, an obligate human pathogen, is a sexually transmitted disease that causes consequential worldwide morbidity both in resource-abundant and resource-limited nations, and its diagnosis and treatment require costly expenditures annually.
Neisseria gonorrhoeae is the second most common bacterial cause of sexually transmitted infections (STIs) in North America, following Chlamydia trachomatis. 1 - 3 Globally, gonococcal infections are now an urgent problem because N. gonorrhoeae is capable of rapidly developing resistance to multiple antibiotic classes. 4 - 8 Over time, N. gonorrh...
Research to elucidate the specific or nonspecific antigens and mechanisms involved in the MeNZB-mediated protection against gonorrhoea is crucial.
INTRODUCTION. Neisseria gonorrhoeae (the gonococcus) is a Gram-negative diplococcus, an obligate human pathogen, and the etiologic agent of the sexually transmitted disease, gonorrhea. The gonococcus infects a diverse array of mucosal surfaces, some of which include the urethra, the endocervix, the pharynx, conjunctiva and the rectum 1.
Gonorrhea is the second most common bacterial sexually transmitted infection in the United States. Rates are increasing, and multiple challenges compound management, including worsening antimicrobial resistance.
Neisseria gonorrhoeae, an obligate human pathogen, is a sexually transmitted disease that causes consequential worldwide morbidity both in resource-abundant and resource-limited nations, and its diagnosis and treatment require costly expenditures annually.
Gonorrhoea is a major public health concern globally. Increasing incidence and sporadic ceftriaxone-resistant cases, including treatment failures, are growing concerns. The 2020 European gonorrhoea guideline provides up-to-date evidence-based guidance regarding the diagnosis and treatment of gonorrhoea. The updates and recommendations emphasize ...
This paper summarizes the evidence discussed at the meeting and the rationale for the 2015 CDC Sexually Transmitted Diseases (STD) treatment guidelines for gonococcal infections in adolescents and adults.
The bacterium Neisseria gonorrhoeae causes the sexually transmitted infection (STI) gonorrhoea, which has an estimated global annual incidence of 86.9 million adults. Gonorrhoea can present as...