• No results found

Neisseria gonorrhoeae: testing, typing and treatment in an era of increased antimicrobial resistance - Chapter 9: General discussion

N/A
N/A
Protected

Academic year: 2021

Share "Neisseria gonorrhoeae: testing, typing and treatment in an era of increased antimicrobial resistance - Chapter 9: General discussion"

Copied!
20
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Neisseria gonorrhoeae: testing, typing and treatment in an era of increased

antimicrobial resistance

Wind, C.M.

Publication date

2017

Document Version

Other version

License

Other

Link to publication

Citation for published version (APA):

Wind, C. M. (2017). Neisseria gonorrhoeae: testing, typing and treatment in an era of

increased antimicrobial resistance.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)
(3)
(4)

9

Steadily, antimicrobial resistance (AMR) in Neisseria gonorrhoeae has emerged for all types of drugs that have been used as fi rst-line treatment.1 As mentioned in

the introduction of this thesis, AMR to either component of the internationally recommended dual therapy, consisting of azithromycin and ceftriaxone, has been reported since 2011.2-14 An outbreak of high-level azithromycin resistance has been

observed in the United Kingdom since 2015,15 which was followed in 2016 by the fi rst

report of dual therapy treatment failure.16 Because no new antimicrobials are expected

shortly, these trends threaten the future treatment of patients with gonorrhoea.17

The fi rst reports of ceftriaxone resistance prompted the World Health Organization (WHO) to publish an action plan with recommendations to stop resistance in gonorrhoea.18 The main suggested strategies included: effective diagnosis and

systematic monitoring of treatment failures, strengthened AMR surveillance, improved networks of laboratories to perform culture, molecular methods to detect AMR, and new treatment strategies. With the studies in this thesis we aimed to provide knowledge on three main topics recommended by the WHO: 1) the improvement of diagnostics for N. gonorrhoeae, 2) surveillance of AMR, and 3) the improvement of treatment strategies. In this last chapter, the main conclusions of the studies in this thesis are compared to existing literature. The implications are discussed, and recommendations for further steps in research are provided.

Improvement of diagnostics for N. gonorrhoeae

In most settings gonorrhoea is diagnosed using a syndromic approach based on symptoms, culture of N. gonorrhoeae, or a nucleic acid amplifi cation test (NAAT).19- 22

Currently, the only way to determine resistance in N. gonorrhoeae is to culture the bacteria and expose them to drugs in vitro.20 Until molecular methods to determine

AMR are widely available, and reliable, successful culture techniques are essential to monitor resistance and individualize treatment. The use of culture is complicated by the required logistics and technical expertise.20 Many low-income countries and

remote settings do not have either, and therefore culture results, and the subsequent antimicrobial susceptibility data are very limited.23 Yet, even in high-income countries

the logistic distance between collection site (an STI or general practitioners clinic) and laboratory creates diffi culties for successful cultures. To improve success rates of culture and allow time for transport of samples, a medium is needed that keeps the sample viable during storage. Several previous studies have shown that the ESwab

(5)

medium could be used for this purpose, but none used clinical samples to evaluate this.24-26

In Chapter 2 we demonstrated a method of targeted deferred culture from the Eswab medium, which was stored for up to 3 days. After selection of samples positive for N.

gonorrhoeae in NAAT, culture success rates were 69% and 56%, after storage for 1 and 2

days, respectively. After storage for 3 days urogenital samples (cervical and urine) still resulted in 44% successful cultures, compared to 22% for rectal samples. These results not only show that N. gonorrhoeae samples can be stored and refrigerated prior to culturing, but also that culture from urine samples is successful. All three procedures were previously thought to be detrimental for N. gonorrhoeae cultures.20,27,28 The

method we presented will enable care providers to transport samples from remote locations to laboratory facilities. By obtaining culture and resistance data, these settings could improve their diagnostic methods, individualize treatment if resistance increases, and contribute to AMR surveillance in their population. Our method of targeted deferred culture could also improve diagnostic procedures in settings with fully equipped laboratories on site. Selection of stored samples for culture, based on NAAT results (potentially available within 24 hours), can avoid many unsuccessful cultures, and improve cost-effectiveness of gonorrhoea diagnostics.

To implement this method in routine diagnostics and AMR surveillance, further studies should be performed to confirm our results in routine practice. These studies should include analyses of the transport times, and success rates of deferred culture from samples collected by general practitioners, in remote settings, or in low-income countries. Another aim should be to perform NAAT and culture from the same medium, allowing care providers to obtain only one sample from patients, and limiting discomfort to patients.

Before the use of extended-spectrum cephalosporins (ESC), treatment failures caused by antimicrobial resistance could be diagnosed by performing a test of cure (TOC). A TOC consisted usually of a culture obtained around 1–2 weeks after treatment, and a negative result was interpreted as a cured infection.20,29,30 This method was complicated by imperfect

sensitivity (around 80%), and false-negative results of cultures.31 When treatment with

ceftriaxone was commenced, and based on its high efficacy, TOC were no longer deemed useful.32,33 In addition, diagnostics for gonorrhoea had improved and were now performed

using high-sensitivity NAATs.29,30 With the looming AMR to ceftriaxone, a TOC could again

(6)

9

In Chapter 3 we analysed the appropriate timing of a TOC for N. gonorrhoeae, using modern ribonucleic acid (RNA)- and deoxyribonucleic acid (DNA)-based NAATs and daily sampling up to 28 days following routine treatment. The median time to clearance was 2 days, and all patients had cleared RNA within 7 days, and DNA within 15 days. The few previous studies, that used either different molecular tests or single time point testing, showed a median time to clearance of 2 days, or complete clearance between 14 and 21 days.34-37 Despite the use of different tests or frequency of

testing in these studies, their results are in line with those of Chapter 3. Our results also confi rm previous reports of intermittent NAAT-positive results in 18% of gonorrhoea patients after treatment.34 Although we used a different molecular test method, we

noted these ‘blips’ in 0.8% and 1.5% (RNA and DNA, respectively) of post-clearance samples. This corresponded to 10% and 16% of patients in our study, respectively. Because NAAT only determines the presence of genetic material, positive results could represent viable infections, shedding of dead bacteria, or the deposition of genetic material. We did not consider blips as reinfections, because positive results were often solitary, not persistent, and not positive for both RNA and DNA. We also noted that test levels (relative light units or cycle threshold) of blips were signifi cantly lower compared to pretreatment samples, suggesting lower genetic load in blips. Some blips were preceded by sexual contact, which suggests that these blips could represent depositions by a partner, although power was too small for statistical analysis. Currently, a TOC is not recommended for anogenital gonorrhoea in routine practice, but this could change if resistance to azithromycin and ceftriaxone continues to increase.20,21,29,30 If a TOC is indicated, we recommend performing it at least 1 or 2 weeks

after treatment, when using RNA- or DNA-based NAAT respectively. Our results suggested faster clearance in rectal samples, or in those treated with ceftriaxone plus doxycycline, but these differences were not statistically signifi cant, possibly due to insuffi cient sample size.

Because this study is the fi rst using modern NAATs and daily testing, our results should be confi rmed by other studies, preferably including pharyngeal infections, and different commercial types of NAAT. Future studies should have a larger sample size, thereby providing more power to analyse associations of clearance with anatomical site or treatment regime, and of blips and sexual contact. A main focus of future studies should be to determine the origin and meaning of blips, confi rming if blips occur, how often, and for how long after treatment. Preferably, a test should be

(7)

designed to establish if test results represent viable or dead bacteria, for instance by testing messenger RNA (mRNA). Until further research sheds more light on the origin of blips, we recommend that a positive TOC should not immediately be interpreted as a treatment failure, but is confirmed with a second sample. Especially if no explanation for reinfection exists, will this help to differentiate between a blip and a treatment failure. When a TOC result suggests a treatment failure, it is important to obtain a culture and determine AMR for the infecting strain.

Coinfections of both N. gonorrhoeae and Chlamydia trachomatis occur in 2–5% of patients at high-risk of having an STI.38-42 Among patients with gonorrhoea, the percentage of

coinfections with chlamydia is much higher at 10–40%.21 Therefore, most clinics test for

both infections simultaneously, and many NAATs give results for both N. gonorrhoeae and C. trachomatis from a single test. While AMR has been a longstanding problem for N. gonorrhoeae, this has not been reported for C. trachomatis. However, there is increasing concern about the effectiveness of azithromycin as first-line treatment in urogenital (and often rectal) chlamydia. Treatment failures of azithromycin for urogenital and rectal chlamydia are occasionally seen, and treatment efficacy is reported to be between 83–97%.43-47 A TOC is currently not routinely recommended for

C. trachomatis, but suggested to be performed 3–4 weeks after treatment, for specific

indications.30,48 However, if efficacy of azithromycin decreases further in the future, a

TOC could be helpful to identify patients with treatment failures.

In Chapter 4 we analysed the NAAT results of a subpopulation from Chapter 3, coinfected with C. trachomatis and N. gonorrhoeae. The median time to clearance of

C. trachomatis was 7 days (RNA) or 6 days (DNA), and all patients cleared within 13

days (RNA) or 15 days (DNA). These results are in line with previous studies showing clearance within 3–4 weeks, although these studies used different molecular tests or frequency of sampling.49,50 However, some reports describe persistence or intermittent

positivity of C. trachomatis up to 51 days.51,52 Because in those studies testing was

not daily, it is unclear whether positive test results represent persistent infection, reinfection, or intermittent positive results (blips) without clinical significance. In some studies 5–18% of patients had positive results preceded by negative results, suggesting that these represent blips.50-54 This is similar to our results of 2% and 4%

blips in post-clearance samples, representing 22% and 35% of patients (DNA and RNA, respectively). However, because we performed daily sampling, we could exclude that chlamydia reinfections occurred. Since blips in our study were preceded and followed by a large number of negative test result, persistent infections were also less likely.

(8)

9

Even though our results are similar to previous studies, and add new knowledge by using modern RNA- and DNA-based NAATs and daily sampling, the sample size of our study was small. Future studies aimed at confi rming our results need to have a larger sample size, and include monoinfections with C. trachomatis. It is also important for future studies to assess possible associations between clearance and anatomical site, human immunodefi ciency virus (HIV) status, or treatment regime (azithromycin or doxycycline), and between blips and sexual contact, coinfections or immune status. Because C. trachomatis is an intracellular growing organism, it is important that the origin of blips is studied in more detail. Especially whether blips represent viable or dead bacteria, and if they are caused by genetic, but non-viable, deposits from a sex partner, or are the result of shedding in the patient. The viability of bacteria could be determined by testing for mRNA. Currently such tests are not yet available, but studies are being performed in this fi eld. Furthermore, it is important to affi rm the effectiveness of azithromycin and doxycycline in anogenital chlamydia, by double blind randomized controlled trials. If indicated, we recommend performing TOC for chlamydia at least 2 weeks after treatment. If a TOC is positive, and a reinfection is unlikely, it needs to be confi rmed with a second sample.

Surveillance of antimicrobial resistance in N. gonorrhoeae

As mentioned before, in recent years AMR to ceftriaxone and/or azithromycin has increased12,17 Most worrying is the fi rst reported treatment failure to dual therapy

of azithromycin and ceftriaxone.16 In Chapter 5 we determined susceptibility to

azithromycin and ceftriaxone in N. gonorrhoeae isolates from patients of the STI Clinic Amsterdam, between 2012 and 2015. We found no resistance to ceftriaxone in Amsterdam yet. Resistance to azithromycin was stable at around 1.2%, while this was 5% across Europe in 2013.12 Although the lower resistance in Amsterdam is a fortunate

situation, we did see a rise in isolates with decreased susceptibility to azithromycin (from 4% in 2012, to 9% in 2015), or to ceftriaxone (from 4% in 2012, to 8% in 2015). This rise of decreased susceptibility was signifi cantly associated with more recent year of infection for both azithromycin and ceftriaxone in MSM, but only for ceftriaxone in heterosexuals. Among heterosexuals, decreased susceptibility to either drug was associated with high-risk behaviour, i.e. by more sex partners in the previous 6 months. Our results confi rm the need for continued monitoring of AMR. Many countries have already established national gonococcal AMR surveillance programs, but in others this needs to be set up as well.22,55-60 It is important that methods and results of surveillance

(9)

the testing methods can differ per county, and sometimes per laboratory. For instance, two different growth media are used (Mueller-Hinton and GC chocolate agar), antimicrobial susceptibility can be determined with agar dilution or Etest methods, and resistance breakpoints used in Europe are not identical to those used in the United States.61-63 For the future it is very important that surveillance is improved, increased

and standardised. The effort to establish national surveillance programs in developing countries needs to be continued and expanded. In countries with strong first-line care such as the Netherlands, AMR surveillance should not be limited to STI clinics, but also include data from general practitioners and outreach programs. Future research should aim at continued reporting of surveillance data, and include essential patient characteristics such as age, gender, sexual orientation, HIV status, and data on sexual risk behaviour. These data are necessary to asses which patients are at highest risk for resistance. Future studies could also help to determine which methods to collect, culture, and determine resistance are most suitable to use for surveillance.

Resistance to azithromycin has been strongly associated with specific genetic mutations. As a result of mutations in any of the four 23S rRNA genes, azithromycin can no longer bind effectively to 23S rRNA, and its inhibitory effect on protein synthesis is limited.64,65 Mutations at position 2059 or 2058 are associated with

high-level azithromycin resistance, while mutations at position 2611 are associated with moderate-level resistance.14,65,66 Furthermore, mutations in one allele are associated

with the rapid introduction of mutations in other alleles, and the cumulative number of C2611T mutated alleles is associated with higher MICs.64,65 Specific strains have also

been associated with resistance, as demonstrated by the occurrence of ESC resistance in sequence type (ST) ST1407 strains from different geographical regions.5,10,67-70

In Chapter 8 we showed that 90% of azithromycin resistant N. gonorrhoeae isolates had at least one C2611T mutation, while all susceptible isolates had a non-mutated wild-type 23S rRNA; this difference was highly significant. These results confirm the previously reported association between azithromycin resistance and 23S rRNA mutations.14,64-66,71 Using N. gonorrhoeae multilocus variable-number tandem

repeat analysis (NG-MLVA), we noted that resistant isolates were significantly more often included in any NG-MLVA hierarchical cluster, and three of the five NG-MLVA clusters consisted predominantly of resistant isolates. This could indicate that these clusters represent sexual networks in which resistant isolates are transmitted. However, four clusters included both resistant and susceptible isolates, suggesting

(10)

9

that resistance occurs independently from NG-MLVA type. NG-MLVA is an in-house developed PCR method that is not widely used, which complicates the comparison of results to those of other studies.72,73 A more commonly used typing method is N.

gonorrhoeae multiantigen sequence typing (NG-MAST).74 Previous studies have linked

azithromycin resistance (among others) to NG-MAST genogroups G2992, G2400 and G1407.13,14,71,75 No correlation has been reported for G5108 or G359.14,71,76-78 In Chapter 8,

azithromycin resistance was signifi cantly associated with genogroups G2992, G5108, and G359, whereas G2400 was signifi cantly associated with susceptibility. We found no association of G1407 with either resistance or susceptibility. Our results, and those from previous studies, suggest that common STs differ by geographical region, and that azithromycin resistance develops independently from the ‘background’ genetic profi le, but that such resistant strains can subsequently spread clonally.

This again stresses the need for continued and improved regional AMR surveillance, which should include epidemiological characteristics to identify risk groups. The association between azithromycin resistance and 23S rRNA mutations allows for fast and targeted analyses of resistance. Other molecular markers are associated with resistance to other antimicrobials. For instance, PenA mosaic types are associated with ESC resistance, and gyrA mutations with ciprofloxacin resistance.72,79-84 However,

these associations are not always very strong, and for many antimicrobials molecular markers associated with resistance have not yet been described.85 Future studies could

help to identify molecular markers, or a combination of markers, in strong association with resistance to different antimicrobial drugs. Combining this knowledge with improving techniques such as whole genome sequencing (WGS) could improve the quality and effi ciency of AMR surveillance in gonorrhoea.14,76,86 When high-defi nition

methods like WGS are used for genetic linkage studies the discriminatory power should be taken into consideration. Too much discriminatory power could limit the possibility to identify clusters and determine risk groups. Another important factor is that many current molecular methods require large quantities of DNA, that are only obtainable after culture. It would be a great improvement to be able to perform cluster analysis on samples directly derived from patients, for instance from NAAT samples, without the need for culture. The determination of AMR by molecular markers also still requires cultured strains. Even when molecular AMR markers will be determined and available for routine testing in the future, this will never cover new mutations that occur spontaneously in the population. Genetic markers of resistance to new gonorrhoea treatment strategies will require cultured strains, that are exposed to antimicrobial

(11)

drugs, to detect the phenotypic AMR. To allow rapid AMR determination, molecular methods need to be improved and expanded, but culture should not be discarded from AMR surveillance

Improvement of treatment strategies in N. gonorrhoeae

To overcome AMR and subsequent treatment failures, various strategies have been used in the past. Treatment options proved effective for a while, but eventually AMR against the new regime emerged, leading again to treatment failures. This was followed by steps to increase the antimicrobial dosage, and then to switch from antimicrobial class. Currently, dual therapy consisting of ceftriaxone plus azithromycin is recommended.17,21,30 Several reasons were put forward to motivate dual therapy.

First, in chronic infections such as human immunodeficiency virus (HIV), leprosy and tuberculosis, combination therapy is effective against AMR, because it is harder for a pathogen to develop resistance to multiple drugs with different modes of action at once. Second, if the bacterial strain is resistant to only one of the components of dual therapy, it will still be treated effectively by the other component.30 Third, gonorrhoea

often coincides with chlamydia, for which the preferred treatment is azithromycin.30,48,87

Therefore the current dual therapy could also treat a possible chlamydia coinfection. Finally, some drug combinations have shown to act synergistically; each drug enhancing the effect of the other, and the combination is more effective than the mere sum of both effects. This effect had been reported for ceftriaxone and azithromycin in N. gonorrhoeae in one study prior to the recommendation of this dual therapy.88

However, in Chapter 6 we found different results.

Using various strains and two testing methods, we determined that the combined effect of ceftriaxone and azithromycin was indifferent, meaning that there was no synergy. However, there was no antagonism either, indicating that the combined effect was not less than the effects of monotherapy. Several other studies have since confirmed our results, refuting the hypothesis that synergy could be a justification for using ceftriaxone and azithromycin.89-92 We also tested 65 other antimicrobial dual

combinations, and found no synergy in any of them. For some of the combinations we tested, and several new combinations, the lack of synergy was confirmed by other in vitro studies.89-92 However, we did identify in vitro efficacy for several existing

antibiotics, that have not yet been first-line choices for gonorrhoea, such as ertapenem, fosfomycin and gentamicin. These drugs are good candidates for future therapy in gonorrhoea, and need to be evaluated further in clinical trials.

(12)

9

Although the lack of synergy between ceftriaxone and azithromycin has been confi rmed by several studies, and resistance to either drug is increasing, the addition of azithromycin is still recommended by international guidelines.29,30 Azithromycin

is widely used; not only for chlamydia, but also for the syndromic management of urethritis, and for many respiratory and cutaneous infections.30,93,94 Therefore,

the population exposure to azithromycin is high, especially in patients at risk of N.

gonorrhoeae infections. Exposure to antimicrobial drugs is the strongest risk factor to

develop resistance.95 The correlation between exposure to macrolides and resistance

has been demonstrated for azithromycin and erythromycin in different micro-organisms.96-98

In Chapter 7 we showed that N. gonorrhoeae isolates from patients who were treated with azithromycin in the 30 days before diagnosis, had minimum inhibitory concentrations (MICs) that were 2.7 times higher than those of strains isolated from patients who had not been treated with azithromycin. This effect was signifi cant, also when adjusted for year of infection, age, ethnic origin, and anatomical site of infection. These results suggest that exposure to azithromycin induces or selects for azithromycin resistance in N. gonorrhoeae. The causative factors for this effect of exposure on MIC remain unresolved. We performed WGS for a subset of samples and noted signifi cantly more mtrR A39T and G45D mutations in those recently exposed. Despite the low number of resistant isolates in our study, these mtrR mutations could possibly explain the increase in MIC. Future studies are needed to determine the effect of exposure to azithromycin on resistance, and the mechanisms by which this effect is achieved. Whether exposure causes selection of resistant isolates that are already present, or if exposure induces resistance in susceptible strains needs to be elucidated. Especially the influence of exposure on 23S rRNA and on mtrR mutations needs to be examined, as well as the effect of mtrR mutations on azithromycin MICs.

The lack of synergy between ceftriaxone and azithromycin, the long half-life of azithromycin, and the evidence that exposure to azithromycin could induce resistance, calls for a re-evaluation of the use of azithromycin in dual therapy for gonorrhoea.99

Future studies should focus on in vitro identifi cation of antimicrobial drugs that are effective against N. gonorrhoeae, and on the correlation between in vitro susceptibility and clinical effect. Antimicrobial drugs with promising in vitro results should be clinically evaluated as soon as possible. Several trials of currently existing drugs have recently been completed, showing clinical effect of gentamicin, gemifloxacin

(13)

and fosfomycin on N. gonorrhoeae.100,101 Randomized controlled trials should first

evaluate promising antimicrobials as monotherapy to establish their effectiveness and adverse events, and only subsequently evaluate these in combination with other drugs. In addition to evaluating existing treatments,102 it is very important that new

antimicrobial drugs are developed and tested for gonorrhoea.

Several new drugs, such as modithromycin, delafloxacin, and a new fluoroquinolone (WQ3810), are being investigated for in vitro efficacy in N. gonorrhoeae.103-105 Currently,

only three new drugs have progressed from in vitro studies to clinical evaluations. The oral ETX0914 (AZD0914) is a new class antimicrobial drug, acting as a DNA gyrase/ topoisomerase II inhibitor. Promising in vitro results have been reported by several studies,106-111 and a phase 1 clinical study in human volunteers showed favourable

pharmacokinetic and -dynamic profiles, whereas adverse events were mild.112 The

results of a completed open-label phase 2 study in patients with gonorrhoea have not yet been published (clinicaltrials.gov identifier: NCT02257918). This is similar to the progress of gepotidacin; after promising in vitro results,113 an open-label phase 2

study is now completed (NCT02294682). The development of solithromycin, a new fluoroketolide, is most advanced. After showing promising in vitro results,114,115 and

good clinical effectiveness in a phase 2 study,116 an open-label phase 3 trial is currently

ongoing (NCT02210325).

Because N. gonorrhoeae is such a fast changing organism, it is to be expected that AMR will occur in the future for new types of drugs. A vaccine reducing or preventing infections in high-risk populations, might prove more effective in stopping the spread of gonorrhoea without causing the induction of AMR. Several bacterial antigens have been identified as possible targets for a vaccine,117-120 enhanced by the use of

proteome mining,121 but this is complicated by the variability of gonococcal surface

antigens.119,121-123 A transgenic mouse-model is also available to test potential vaccines,

but clinical tests in humans have so far been unsuccessful.119,121,122 The lack of an innate

immune response after infections with N. gonorrhoeae, and insufficient knowledge of protective immune responses further complicate vaccine development.119,122,123

Concluding remarks

Based on previous research and the results of studies in this thesis it is to be expected that antimicrobial resistance in N. gonorrhoeae will further increase in the near future. To keep up with, and preferably stay ahead of, this development it is crucial that

(14)

9

AMR surveillance is intensifi ed across the world. Surveillance should focus on the identifi cation of molecular markers for resistance, and rapid, effective test methods to determine their presence in circulating strains. However, while molecular methods are increasingly used, the knowledge and implication of culture should not be abandoned. Research on alternative treatment options should focus on three topics: reappraisal of existing antimicrobial drugs and their effectiveness against N. gonorrhoeae, determined both in vitro and in clinical trials, the development of new antimicrobial drugs, and the development of a vaccine for high-risk groups. This requires laboratory techniques, time, and effort. Moreover, the required funding for large studies and the development of new techniques and treatments is absolutely indispensable. By improving international collaborations, and gaining the attention of governments, the funding for AMR research in N. gonorrhoeae could be improved.

Before novel treatment strategies are available, we could already improve the care of patients with gonorrhoea by focussing on individualized treatment. By using culture or molecular testing, we could identify strains that are still susceptible to previously used antibiotics, such as penicillin, spectinomycin, or ciprofloxacin, and treat patients accordingly. Finally, education of high-risk populations, and continued promotion of condom use could prevent many infections, and thereby limit the spread of antimicrobial resistance.

(15)

REFERENCES

1. 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.

2. Ohnishi M, Saika T, Hoshina S, et al. Ceftriaxone-resistant Neisseria gonorrhoeae, Japan. Emerg Infect Dis. 2011;17(1):148-149.

3. 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(8):1858-1860.

4. Martin I, Sawatzky P, Allen V, et al. Emergence and characterization of Neisseria gonorrhoeae isolates with decreased susceptibilities to ceftriaxone and cefixime in Canada: 2001-2010. Sex Transm Dis. 2012;39(4):316-323.

5. 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(3):1273-1280.

6. 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(25):pii:20200.

7. 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(6):1267-1270.

8. 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(6):841-843.

9. Bercot B, Belkacem A, Goubard A, et al. High-level azithromycin-resistant Neisseria gonorrhoeae clinical isolate

in France, March 2014. Euro Surveill. 2014;19(44):pii:20951.

10. Morita-Ishihara T, Unemo M, Furubayashi K, et al. Treatment failure with 2 g of azithromycin (extended-release formulation) in gonorrhoea in Japan caused by the international multidrug-resistant ST1407 strain of Neisseria gonorrhoeae. J Antimicrob Chemother. 2014;69(8):2086-2090.

11. Lynagh Y, Mac Aogain M, Walsh A, Rogers TR, Unemo M, Crowley B. Detailed characterization of the first high-level azithromycin-resistant Neisseria gonorrhoeae cases in Ireland. J Antimicrob Chemother. 2015;70(8):2411-2413.

12. Cole MJ, Spiteri G, Jacobsson S, et al. Is the tide turning again for cephalosporin resistance in Neisseria

gonorrhoeae in Europe? Results from the 2013 European surveillance. BMC Infect Dis. 2015;15:321.

13. Brunner A, Nemes-Nikodem E, Jeney C, et al. Emerging azithromycin-resistance among the Neisseria

gonorrhoeae strains isolated in Hungary. Ann Clin Microbiol Antimicrob. 2016;15(1):53.

14. Jacobsson S, Golparian D, Cole M, 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. 2016; pii: dkw279. Epub ahead of print.

15. Chisholm SA, Wilson J, Alexander S, et al. An outbreak of high-level azithromycin resistant Neisseria

gonorrhoeae in England. Sex Transm Infect. 2016;92(5):365-367.

16. Fifer H, Natarajan U, Jones L, et al. Failure of dual antimicrobial therapy in treatment of gonorrhea. N Engl J Med. 2016;374(25):2504-2506.

17. Unemo M, Del Rio C, Shafer WM. Antimicrobial resistance expressed by Neisseria gonorrhoeae: a major global public health problem in the 21st Century. Microbiol Spectr. 2016;4(3):doi:10.1128/microbiolspec.EI1110-0009-2015.

18. World Health Organization. Global action plan to control the spread and impact of antimicrobial resistance in Neisseria gonorrhoeae. Geneva: WHO;2012.

19. Yin YP, Wu Z, Lin C, et al. Syndromic and laboratory diagnosis of sexually transmitted infection: a comparative study in China. Int J STD AIDS. 2008;19(6):381-384.

20. Centers for Disease Control and Prevention. Recommendations for the laboratory-based detection of

Chlamydia trachomatis and Neisseria gonorrhoeae – 2014. MMWR Recomm Rep. 2014;63(RR02):1-19.

(16)

9

22. Hananta IP, van Dam AP, Bruisten SM, Schim van der Loeff MF, 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(10):608-616.

23. Ndowa FJ, Francis JM, Machiha A, Faye-Kette H, Fonkoua MC. Gonococcal antimicrobial resistance: perspectives from the African region. Sex Transm Infect. 2013;89 Suppl 4:iv11-15.

24. Drake C, Barenfanger J, Lawhorn J, Verhulst S. Comparison of Easy-Flow Copan Liquid Stuart’s and Starplex Swab transport systems for recovery of fastidious aerobic bacteria. J Clin Microbiol. 2005;43(3):1301-1303. 25. Rishmawi N, Ghneim R, Kattan R, et al. Survival of fastidious and nonfastidious aerobic bacteria in three

bacterial transport swab systems. J Clin Microbiol. 2007;45(4):1278-1283.

26. Van Horn KG, Audette CD, Sebeck D, Tucker KA. Comparison of the Copan ESwab system with two Amies agar swab transport systems for maintenance of microorganism viability. J Clin Microbiol. 2008;46(5):1655-1658. 27. Sng EH, Rajan VS, Yeo KL, Goh AJ. The recovery of Neisseria gonorrhoeae from clinical specimens: effects of

different temperatures, transport times, and media. Sex Transm Dis. 1982;9(2):74-78.

28. Rosey CE, Britt EM. Urine as a holding medium for Neisseria gonorrhoeae. Sex Transm Dis. 1984;11(4):301-303. 29. 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.

30. Centers for Disease Control and Prevention. Sexually transmitted diseases treatment guidelines, 2015. MMWR Recomm Rep. 2015;64(RR-03):1-137.

31. Mohammed H, Ison CA, Obi C, et al. Frequency and correlates of culture-positive infection with Neisseria

gonorrhoeae in England: a review of sentinel surveillance data. Sex Transm Infect. 2015;91(4):287-293.

32. Harry C. The management of uncomplicated adult gonococcal infection: should test of cure still be routine in patients attending genitourinary medicine clinics? Int J STD AIDS. 2004;15(7):453-458.

33. Centers for Disease Control and Prevention. Sexually transmitted diseases treatment guidelines, 2010. MMWR Recomm Rep. 2010;59(RR-12):1-116.

34. Bachmann LH, Desmond RA, Stephens J, Hughes A, Hook EW, 3rd. 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(10):3596-3601.

35. Hjelmevoll SO, Olsen ME, Sollid JU, et al. Appropriate time for test-of-cure when diagnosing gonorrhoea with a nucleic acid amplifi cation test. Acta Derm Venereol. 2012;92(3):316-319.

36. 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(10):595-600. 37. Bissessor M, Whiley DM, Fairley CK, et al. Persistence of Neisseria gonorrhoeae DNA following treatment for

pharyngeal and rectal gonorrhea is influenced by antibiotic susceptibility and reinfection. Clin Infect Dis. 2015;60(4):557-563.

38. Tongtoyai J, Todd CS, Chonwattana W, 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(8):440-449.

39. Guy R, Ward J, Wand H, et al. Coinfection with Chlamydia trachomatis, Neisseria gonorrhoeae and Trichomonas

vaginalis: a cross-sectional analysis of positivity and risk factors in remote Australian Aboriginal communities.

Sex Transm Infect. 2015;91(3):201-206.

40. 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(9):e007691.

41. Dubbink JH, de Waaij DJ, Bos M, et al. Microbiological characteristics of Chlamydia trachomatis and Neisseria

gonorrhoeae infections in South African women. J Clin Microbiol. 2016;54(1):200-203.

42. Tao G, Hoover KW, Nye MB, et al. Rectal infection with Neisseria gonorrhoeae and Chlamydia trachomatis in men in the United States. Clin Infect Dis. 2016;63(10):1325-1331.

43. Geisler WM, Uniyal A, Lee JY, et al. Azithromycin versus doxycycline for urogenital Chlamydia trachomatis infection. N Engl J Med. 2015;373(26):2512-2521.

44. Kong FY, Tabrizi SN, Fairley CK, et al. The effi cacy of azithromycin and doxycycline for the treatment of rectal chlamydia infection: a systematic review and meta-analysis. J Antimicrob Chemother. 2015;70(5):1290-1297. 45. Kong FY, Tabrizi SN, Fairley CK, et al. Higher organism load associated with failure of azithromycin to treat

(17)

46. Gratrix J, Brandley J, Dane M, et al. A retrospective review of treatment failures using azithromycin and doxycycline in the treatment of rectal chlamydia infections in women and men who have sex with men. Sex Transm Dis. 2016;43(2):110-112.

47. Kissinger PJ, White S, Manhart LE, et al. Azithromycin treatment failure for Chlamydia trachomatis among heterosexual men with nongonococcal urethritis. Sex Transm Dis. 2016;43(10):599-602.

48. World Health Organization. WHO guidelines for the treatment of Chlamydia trachomatis. Geneva: WHO;2016. 49. Claas HC, Wagenvoort JH, Niesters HG, Tio TT, Van Rijsoort-Vos JH, Quint WG. Diagnostic value of the

polymerase chain reaction for Chlamydia detection as determined in a follow-up study. J Clin Microbiol. 1991;29(1):42-45.

50. Gaydos CA, Crotchfelt KA, Howell MR, Kralian S, Hauptman P, Quinn TC. Molecular amplification assays to detect chlamydial infections in urine specimens from high school female students and to monitor the persistence of chlamydial DNA after therapy. J Infect Dis. 1998;177(2):417-424.

51. Dukers-Muijrers NH, Morre SA, Speksnijder A, van der Sande MA, Hoebe CJ. Chlamydia trachomatis test-of-cure cannot be based on a single highly sensitive laboratory test taken at least 3 weeks after treatment. PLoS One. 2012;7(3):e34108.

52. Sena AC, Lensing S, Rompalo A, et al. Chlamydia trachomatis, Mycoplasma genitalium, and Trichomonas vaginalis infections in men with nongonococcal urethritis: predictors and persistence after therapy. J Infect Dis. 2012;206(3):357-365.

53. Jang D, Sellors J, Howard M, et al. Correlation between culture testing of swabs and ligase chain reaction of first void urine from patients recently treated for Chlamydia trachomatis. Sex Transm Infect. 2003;79(3):237-239.

54. Renault CA, Israelski DM, Levy V, Fujikawa BK, Kellogg TA, Klausner JD. Time to clearance of Chlamydia

trachomatis ribosomal RNA in women treated for chlamydial infection. Sex Health. 2011;8(1):69-73.

55. Koedijk FD, van Veen MG, de Neeling AJ, Linde GB, van der Sande MA. Increasing trend in gonococcal resistance to ciprofloxacin in The Netherlands, 2006-8. Sex Transm Infect. 2010;86(1):41-45.

56. Town K, Obi C, Quaye N, Chisholm S, Hughes G, Group GC. Drifting towards ceftriaxone treatment failure in gonorrhoea: risk factor analysis of data from the Gonococcal Resistance to Antimicrobials Surveillance Programme in England and Wales. Sex Transm Infect. 2016; pii: sextrans-2016-052583. Epub ahead of print. 57. Kirkcaldy RD, Harvey A, Papp JR, et al. Neisseria gonorrhoeae antimicrobial susceptibility surveillance - The

Gonococcal Isolate Surveillance Project, 27 Sites, United States, 2014. MMWR Surveill Summ. 2016;65(7):1-19. 58. Lahra MM, Australian Gonococcal Surveillance P. Australian Gonococcal Surveillance Programme annual

report, 2014. Commun Dis Intell Q Rep. 2015;39(3):E347-354.

59. Hancali A, Ndowa F, Bellaji B, et al. Antimicrobial resistance monitoring in Neisseria gonorrhoeae and strategic use of funds from the Global Fund to set up a systematic Moroccan gonococcal antimicrobial surveillance programme. Sex Transm Infect. 2013;89 Suppl 4:iv24-27.

60. Kubanov A, Vorobyev D, Chestkov A, et al. Molecular epidemiology of drug-resistant Neisseria gonorrhoeae in Russia (Current Status, 2015). BMC Infect Dis. 2016;16:389.

61. Clinical and Laboratory Standards Institute. Performance standards for antimicrobial susceptibility testing; twenty-second informational supplement. In: Document M100-S22. Wayne, PA: CLSI;2012.

62. European Committee on Antimicrobial Susceptibility Testing. Breakpoint table for interpretation of MICs and zone diameters, version 6.0. Stockholm: EUCAST;2016.

63. Ison CA, Martin IM, Lowndes CM, Fenton KA, Network E. Comparability of laboratory diagnosis and antimicrobial susceptibility testing of Neisseria gonorrhoeae from reference laboratories in Western Europe. J Antimicrob Chemother. 2006;58(3):580-586.

64. Ng LK, Martin I, Liu G, Bryden L. Mutation in 23S rRNA associated with macrolide resistance in Neisseria

gonorrhoeae. Antimicrob Agents Chemother. 2002;46(9):3020-3025.

65. Chisholm SA, Dave J, Ison CA. High-level azithromycin resistance occurs in Neisseria gonorrhoeae as a result of a single point mutation in the 23S rRNA genes. Antimicrob Agents Chemother. 2010;54(9):3812-3816. 66. Grad YH, Harris SR, Kirkcaldy RD, et al. Genomic epidemiology of gonococcal resistance to extended spectrum

cephalosporins, macrolides, and fluoroquinolones in the US, 2000-2013. J Infect Dis. 2016;214(10):1579-1587. 67. Anselmo A, Ciammaruconi A, Carannante A, et al. Draft genome sequence of Neisseria gonorrhoeae sequence

type 1407, a multidrug-resistant clinical isolate. Genome Announc. 2015;3(4):pii: e00903-15.

68. Tanaka M, Furuya R, Irie S, Kanayama A, Kobayashi I. High prevalence of azithromycin-resistant Neisseria

gonorrhoeae isolates with a multidrug resistance phenotype in Fukuoka, Japan. Sex Transm Dis.

(18)

9

69. Chen MY, Stevens K, Tideman R, et al. Failure of 500 mg of ceftriaxone to eradicate pharyngeal gonorrhoea, Australia. J Antimicrob Chemother. 2013;68(6):1445-1447.

70. Jeverica S, Golparian D, Maticic M, Potocnik M, Mlakar B, Unemo M. Phenotypic and molecular characterization of Neisseria gonorrhoeae isolates from Slovenia, 2006-12: rise and fall of the multidrug-resistant NG-MAST genogroup 1407 clone? J Antimicrob Chemother. 2014;69(6):1517-1525.

71. Belkacem A, Jacquier H, Goubard A, et al. Molecular epidemiology and mechanisms of resistance of azithromycin-resistant Neisseria gonorrhoeae isolated in France during 2013-14. J Antimicrob Chemother. 2016;71(9):2471-2478.

72. Heymans R, Bruisten SM, Golparian D, Unemo M, de Vries HJC, van Dam AP. Clonally related Neisseria

gonorrhoeae isolates with decreased susceptibility to the extended-spectrum cephalosporin cefotaxime in

Amsterdam, the Netherlands. Antimicrob Agents Chemother. 2012;56(3):1516-1522.

73. Heymans R, Schouls LM, van der Heide HG, van der Loeff MF, Bruisten SM. Multiple-locus variable-number tandem repeat analysis of Neisseria gonorrhoeae. J Clin Microbiol. 2011;49(1):354-363.

74. Martin IM, Ison CA, Aanensen DM, Fenton KA, Spratt BG. Rapid sequence-based identifi cation of gonococcal transmission clusters in a large metropolitan area. J Infect Dis. 2004;189(8):1497-1505.

75. Carannante A, Renna G, Dal Conte I, et al. Changing antimicrobial resistance profi les among Neisseria

gonorrhoeae isolates in Italy, 2003 to 2012. Antimicrob Agents Chemother. 2014;58(10):5871-5876.

76. Demczuk W, Martin I, Peterson S, et al. Genomic epidemiology and molecular resistance mechanisms of azithromycin-resistant Neisseria gonorrhoeae in Canada from 1997 to 2014. J Clin Microbiol. 2016;54(5):1304-1313.

77. Liang JY, Cao WL, Li XD, et al. Azithromycin-resistant Neisseria gonorrhoeae isolates in Guangzhou, China (2009-2013): coevolution with decreased susceptibilities to ceftriaxone and genetic characteristics. BMC Infect Dis. 2016;16:152.

78. Shigemura K, Osawa K, Miura M, et al. Azithromycin resistance and its mechanism in Neisseria gonorrhoeae strains in Hyogo, Japan. Antimicrob Agents Chemother. 2015;59(5):2695-2699.

79. Belland RJ, Morrison SG, Ison C, Huang WM. Neisseria gonorrhoeae acquires mutations in analogous regions of gyrA and parC in fluoroquinolone-resistant isolates. Mol Microbiol. 1994;14(2):371-380.

80. Ameyama S, Onodera S, Takahata M, et al. Mosaic-like structure of penicillin-binding protein 2 Gene (penA) in clinical isolates of Neisseria gonorrhoeae with reduced susceptibility to cefi xime. Antimicrob Agents Chemother. 2002;46(12):3744-3749.

81. Seike K, Yasuda M, Hatazaki K, et al. Novel penA mutations identifi ed in Neisseria gonorrhoeae with decreased susceptibility to ceftriaxone isolated between 2000 and 2014 in Japan. J Antimicrob Chemother. 2016;71(9):2466-2470.

82. Nakayama S, Shimuta K, Furubayashi K, Kawahata T, Unemo M, Ohnishi M. New ceftriaxone- and multidrug-resistant Neisseria gonorrhoeae strain with a novel mosaic penA gene isolated in Japan. Antimicrob Agents Chemother. 2016;60(7):4339-4341.

83. Hemarajata P, Yang S, Soge OO, Humphries RM, Klausner JD. Performance and verifi cation of a real-time PCR assay targeting the gyrA gene for prediction of ciprofloxacin resistance in Neisseria gonorrhoeae. J Clin Microbiol. 2016;54(3):805-808.

84. Pond MJ, Hall CL, Miari VF, et al. Accurate detection of Neisseria gonorrhoeae ciprofloxacin susceptibility directly from genital and extragenital clinical samples: towards genotype-guided antimicrobial therapy. J Antimicrob Chemother. 2016;71(4):897-902.

85. Ison CA, Golparian D, Saunders P, Chisholm S, Unemo M. Evolution of Neisseria gonorrhoeae is a continuing challenge for molecular detection of gonorrhoea: false negative gonococcal porA mutants are spreading internationally. Sex Transm Infect. 2013;89(3):197-201.

86. Demczuk W, Lynch T, Martin I, et al. Whole-genome phylogenomic heterogeneity of Neisseria gonorrhoeae isolates with decreased cephalosporin susceptibility collected in Canada between 1989 and 2013. J Clin Microbiol. 2015;53(1):191-200.

87. Lanjouw E, Ouburg S, de Vries HJ, Stary A, Radcliffe K, Unemo M. 2015 European guideline on the management of Chlamydia trachomatis infections. Int J STD AIDS. 2016;27(5):333-348.

88. Furuya R, Nakayama H, Kanayama A, et al. In vitro synergistic effects of double combinations of beta-lactams and azithromycin against clinical isolates of Neisseria gonorrhoeae. J Infect Chemother. 2006;12(4):172-176. 89. Barbee LA, Soge OO, Holmes KK, Golden MR. In vitro synergy testing of novel antimicrobial combination

(19)

90. Pereira R, Cole MJ, Ison CA. Combination therapy for gonorrhoea: in vitro synergy testing. J Antimicrob Chemother. 2013;68(3):640-643.

91. Hauser C, Hirzberger L, Unemo M, Furrer H, Endimiani A. In vitro activity of fosfomycin alone and in combination with ceftriaxone or azithromycin against clinical Neisseria gonorrhoeae isolates. Antimicrob Agents Chemother. 2015;59(3):1605-1611.

92. Pettus K, Sharpe S, Papp JR. In vitro assessment of dual drug combinations to inhibit growth of Neisseria

gonorrhoeae. Antimicrob Agents Chemother. 2015;59(4):2443-2445.

93. Horner P, Blee K, O’Mahony C, et al. 2015 UK National Guideline on the management of non-gonococcal urethritis. Int J STD AIDS. 2016;27(2):85-96.

94. Shulman ST, Bisno AL, Clegg HW, et al. Clinical practice guideline for the diagnosis and management of group A streptococcal pharyngitis: 2012 update by the Infectious Diseases Society of America. Clin Infect Dis. 2012;55(10):1279-1282.

95. Fingerhuth SM, Bonhoeffer S, Low N, Althaus CL. Antibiotic-resistant Neisseria gonorrhoeae spread faster with more treatment, not more sexual partners. PLoS Pathog. 2016;12(5):e1005611.

96. King A, Bathgate T, Phillips I. Erythromycin susceptibility of viridans streptococci from the normal throat flora of patients treated with azithromycin or clarithromycin. Clin Microbiol Infect. 2002;8(2):85-92. 97. Young H, Moyes A, McMillan A. Azithromycin and erythromycin resistant Neisseria gonorrhoeae following

treatment with azithromycin. Int J STD AIDS. 1997;8(5):299-302.

98. 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(11):877-879.

99. Horner P, Saunders J. Should azithromycin 1 g be abandoned as a treatment for bacterial STIs? The case for and against. Sex Transm Infect. 2016;pii: sextrans-2015-052414. Epub ahead of print.

100. 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(8):1083-1091.

101. Yuan Z, He C, Yan S, Ke Y, Tang W. Randomized controlled clinical trial on the efficacy of fosfomycin trometamol for uncomplicated gonococcal urethritis in men. Clin Microbiol Infect. 2016;22(6):507-512. 102. Ghathian K, Calum H, Gyssens IC, Frimodt-Moller N. Temocillin in vitro activity against recent clinical isolates

of Neisseria gonorrhoeae compared with penicillin, ceftriaxone and ciprofloxacin. J Antimicrob Chemother. 2016;71(4):1122-1123.

103. Jacobsson S, Golparian D, Phan LT, et al. In vitro activities of the novel bicyclolides modithromycin (EDP-420, EP-013(EDP-420, S-013420) and EDP-322 against MDR clinical Neisseria gonorrhoeae isolates and international reference strains. J Antimicrob Chemother. 2015;70(1):173-177.

104. Kazamori D, Aoi H, Sugimoto K, et al. In vitro activity of WQ-3810, a novel fluoroquinolone, against multidrug-resistant and fluoroquinolone-resistant pathogens. Int J Antimicrob Agents. 2014;44(5):443-449. 105. Soge OO, Salipante SJ, No D, Duffy E, Roberts MC. In vitro activity of delafloxacin against clinical Neisseria

gonorrhoeae isolates and selection of gonococcal delafloxacin resistance. Antimicrob Agents Chemother.

2016;60(5):3106-3111.

106. Alm RA, Lahiri SD, Kutschke A, et al. Characterization of the novel DNA gyrase inhibitor AZD0914: low resistance potential and lack of cross-resistance in Neisseria gonorrhoeae. Antimicrob Agents Chemother. 2015;59(3):1478-1486.

107. Huband MD, Bradford PA, Otterson LG, et al. In vitro antibacterial activity of AZD0914, a new spiropyrimidinetrione DNA gyrase/topoisomerase inhibitor with potent activity against Gram-positive, fastidious Gram-Negative, and atypical bacteria. Antimicrob Agents Chemother. 2015;59(1):467-474. 108. Jacobsson S, Golparian D, Alm RA, 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. 2014;58(9):5585-5588.

109. Kern G, Palmer T, Ehmann DE, et al. Inhibition of Neisseria gonorrhoeae Type II Topoisomerases by the Novel Spiropyrimidinetrione AZD0914. J Biol Chem. 2015;290(34):20984-20994.

110. Unemo M, Ringlander J, Wiggins C, Fredlund H, Jacobsson S, Cole M. High in vitro susceptibility to the novel spiropyrimidinetrione ETX0914 (AZD0914) among 873 contemporary clinical Neisseria gonorrhoeae isolates from 21 European countries from 2012 to 2014. Antimicrob Agents Chemother. 2015;59(9):5220-5225.

(20)

9

111. Su XH, Wang BX, Le WJ, et al. Multidrug-resistant Neisseria gonorrhoeae isolates from Nanjing, China, are sensitive to killing by a novel DNA gyrase inhibitor, ETX0914 (AZD0914). Antimicrob Agents Chemother. 2015;60(1):621-623.

112. Basarab GS, Kern GH, McNulty J, et al. Responding to the challenge of untreatable gonorrhea: ETX0914, a fi rst-in-class agent with a distinct mechanism-of-action against bacterial Type II topoisomerases. Sci Rep. 2015;5:11827.

113. Jones RN, Fedler KA, Scangarella-Oman NE, Ross JE, Flamm RK. Multicenter investigation of gepotidacin (GSK2140944) agar dilution quality control determinations for Neisseria gonorrhoeae ATCC 49226. Antimicrob Agents Chemother. 2016;60(7):4404-4406.

114. 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(5):2739-2742.

115. Riedel S, Ross JE, Farrell DJ, Flamm RK, Jones RN. Determination of disk diffusion and MIC quality control guidelines for solithromycin, a novel fluoroketolide antibacterial, against Neisseria gonorrhoeae. J Clin Microbiol. 2015;53(12):3888-3890.

116. Hook EW, 3rd, Golden M, Jamieson BD, et al. A phase 2 trial of oral solithromycin 1200 mg or 1000 mg as single-dose oral therapy for uncomplicated gonorrhea. Clin Infect Dis. 2015;61(7):1043-1048.

117. Cash DR, Noinaj N, Buchanan SK, Cornelissen CN. Beyond the crystal structure: insight into the function and vaccine potential of TbpA expressed by Neisseria gonorrhoeae. Infect Immun. 2015;83(11):4438-4449. 118. Chakraborti S, Lewis LA, Cox AD, et al. Phase-variable heptose I glycan extensions modulate effi cacy of 2C7

vaccine antibody directed against Neisseria gonorrhoeae lipooligosaccharide. J Immunol. 2016;196(11):4576-4586.

119. Jerse AE, Deal CD. Vaccine research for gonococcal infections: where are we? Sex Transm Infect. 2013;89 Suppl 4:iv63-68.

120. McLaughlin SE, Cheng H, Ghanem KG, et al. Urethral exudates of men with Neisseria gonorrhoeae infections select a restricted lipooligosaccharide phenotype during transmission. J Infect Dis. 2012;206(8):1227-1232. 121. Zielke RA, Wierzbicki IH, Baarda BI, et al. Proteomics-driven antigen discovery for development of vaccines

against gonorrhea. Mol Cell Proteomics. 2016;15(7):2338-2355.

122. Edwards JL, Jennings MP, Apicella MA, Seib KL. Is gonococcal disease preventable? The importance of understanding immunity and pathogenesis in vaccine development. Crit Rev Microbiol. 2016;42(6):928-941. 123. Wetzler LM, Feavers IM, Gray-Owen SD, Jerse AE, Rice PA, Deal CD. Summary and recommendations from

the National Institute of Allergy and Infectious Diseases (NIAID) workshop “Gonorrhea vaccines: the way forward”. Clin Vaccine Immunol. 2016;23(8):656-663.

Referenties

GERELATEERDE DOCUMENTEN

Programming Strategies for Postpartum Family Planning, as well as Jhpiego specific findings from our own PPFP Choices private sector assessments in study sites conducted prior to the

In an incongruent trial, the target appeared on the side of the face with the opposite valence as the context sentence.. Each participant completed 40 congruent trials and

The distinction between direct and indirect effects differentiates between miRNAs that affect the fibrotic response directly by targeting ECM components or fibrotic signalling in

Cardiomyocyte-specific miRNA-30c downregulation through expression of a miRNA-30c- sponge results in strongly decreased miRNA expression (~60% loss) but has no effect on heart

Since miRNA-30c has previously been implicated in the regulation of mitochondrial fission in neonatal cardiomyocytes in vitro, we performed electron microscopy on adult cardiac

On the other hand we studied the in vivo role of miRNA-30c, a miRNA that has been linked to the development of cardiac fibrosis and heart failure.. Chapter 1 serves as an

De relatie tussen archieven en de staat is echter uitermate complex en is gebaseerd op een interessante paradox waarover, zeker door postmoderne wetenschappers, al veel

We performed a two-sided correlational test using the individual values of the connection strength between vmPFC and STN together with the individual values of choice bias (e.g.,