首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
The Zika virus outbreak in the Americas has caused global concern. To help accelerate this fight against Zika, we launched the OpenZika project. OpenZika is an IBM World Community Grid Project that uses distributed computing on millions of computers and Android devices to run docking experiments, in order to dock tens of millions of drug-like compounds against crystal structures and homology models of Zika proteins (and other related flavivirus targets). This will enable the identification of new candidates that can then be tested in vitro, to advance the discovery and development of new antiviral drugs against the Zika virus. The docking data is being made openly accessible so that all members of the global research community can use it to further advance drug discovery studies against Zika and other related flaviviruses.The Zika virus (ZIKV) has emerged as a major public health threat to the Americas as of 2015 [1]. We have previously suggested that it represents an opportunity for scientific collaboration and open scientific exchange [2]. The health of future generations may very well depend on the decisions we make, our willingness to share our findings quickly, and open collaboration to rapidly find a cure for this disease. Since February 1, 2016, when the World Health Organization deemed the cluster of microcephaly cases, Guillain-Barré, and other neurological disorders associated with ZIKV in Latin America and the Caribbean as constituting a Public Health Emergency of International Concern [3] (PHEIC), we have seen a rapid increase in publications (S1 References and main references). We [2] and others [4,5] described steps that could be taken to initiate a drug discovery program on ZIKV. For example, computational approaches, such as virtual screening of chemical libraries or focused screening to repurpose FDA and/or EU-approved drugs, can be used to help accelerate the discovery of an anti-ZIKV drug. An antiviral drug discovery program can be initiated using structure-based design, based on homology models of the key ZIKV proteins. With the lack of structural information regarding the proteins of ZIKV, we built homology models for all the ZIKV proteins, based on close homologs such as dengue virus, using freely available software [6] (S1 Table). These were made available online on March 3, 2016. We also predicted the site of glycosylation of glycoprotein E as Asn154, which was recently experimentally verified [7].Since the end of March 2016, we have now seen two cryo-EM structures and 16 crystal structures of five target classes (S1 Table). These structures, alongside the homology models, represent potential starting points for docking-based virtual screening campaigns to help find molecules that are predicted to have high affinity with ZIKV proteins. These predictions can then be tested against the virus in cell-based assays and/or using individual protein-based assays. There are millions of molecules available that can be assayed, but which ones are likely to work, and how should we prioritize them?In March, we initiated a new open collaborative project called OpenZika (Fig 1), with IBM’s World Community Grid (WCG, worldcommunitygrid.org), which has been used previously for distributed computing projects (S2 Table). On May 18, 2016, the OpenZika project began the virtual screening of ~6 million compounds that are in the ZINC database (Fig 1), as well as the FDA-approved drugs and the NIH clinical collection, using AutoDock Vina and the homology models and crystal structures (S1 Table, S1 Text, S1 References), to discover novel candidate compounds that can potentially be developed into new drugs for treating ZIKV. These will be followed by additional virtual screens with a new ZINC library of ~38 million compounds, and the PubChem database (at most ~90 million compounds), after their structures are prepared for docking.Open in a separate windowFig 1Workflow for the OpenZika project.A. Docking input files of the targets and ligands are prepared, and positive control docking studies are performed. The crystallographic binding mode of a known inhibitor is shown as sticks with dark purple carbon atoms, while the docked binding mode against the NS5 target from HCV has cyan carbons. Our pdbqt files of the libraries of compounds we screen are also openly accessible (http://zinc.docking.org/pdbqt/). B. We have already prepared the docking input files for ~6 million compounds from ZINC (i.e., the libraries that ALP previously used in the GO Fight Against Malaria project on World Community Grid), which are currently being used in the initial set of virtual screens on OpenZika. C. IBM’s World Community Grid is an internet-distributed network of millions of computers (Mac, Windows, and Linux) and Android-based tablets or smartphones in over 80 countries. Over 715,000 volunteers donate their dormant computer time (that would otherwise be wasted) towards different projects that are both (a) run by an academic or nonprofit research institute, and (b) are devoted to benefiting humanity. D. OpenZika is harnessing World Community Grid to dock millions of commercially available compounds against multiple ZIKV homology models and crystal structures (and targets from related viruses) using AutoDock Vina (AD Vina). This ultimately produces candidates (virtual hits that produced the best docking scores and displayed the best interactions with the target during visual inspection) against individual proteins, which can then be prioritized for in vitro testing by collaborators. After it is inspected, all computational data against ZIKV targets will be made open to the public on our website (http://openzika.ufg.br/experiments/#tab-id-7), and OpenZika results are also available upon request. The computational and experimental data produced will be published as quickly as possible.Initially, compounds are being screened against the ZIKV homologs of drug targets that have been well-validated in research against dengue and hepatitis C viruses, such as NS5 and Glycoprotein E (S1 Table, S1 Text, S1 References). These may allow us to identify broad-spectrum antivirals against multiple flaviviruses, such as dengue virus, West Nile virus, and yellow fever virus. In addition, docking against the crystal structure of a related protein from a different pathogen can sometimes discover novel hits against the pathogen of interest [8].As well as applying docking-based filters, the compounds virtually screened on OpenZika will also be filtered using machine learning models (S1 Text, S1 References). These should be useful selection criteria for subsequent tests by our collaborators in whole-cell ZIKV assays, to verify their antiviral activity for blocking ZIKV infection or replication. Since all OpenZika docking data will be in the public domain soon after they are completed and verified, we and other labs can then advance the development of some of these new virtual candidates into experimentally validated hits, leads, and drugs through collaborations with wet labs.This exemplifies open science, which should help scientists around the world as they address the long and arduous process of discovering and developing new drugs. Screening millions of compounds against many different protein models in this way would take far more resources and time than any academic researcher could generally obtain or spend. As of August 16, 2016, we have submitted 894 million docking jobs. Over 6,934 CPU years have been donated to us, enabling over 439 million different docking jobs. We recently selected an initial batch of candidates for NS3 helicase (data openly available at http://openzika.ufg.br/experiments/#tab-id-7), for in vitro testing. Without the unique community of volunteers and tremendous resources provided by World Community Grid, this project would have been very difficult to initiate in a reasonable time frame at this scale.The OpenZika project will ultimately generate several billion docking results, which could make it the largest computational drug discovery project ever performed in academia. The potential challenges we foresee will be finding laboratories with sufficient funding to pursue compounds, synthesize analogs, and develop target-based assays to validate our predictions and generate SAR (Structure-Activity Relationship) data to guide the process of developing the new hits into leads and then drugs. Due to the difficult nature of drug discovery and the eventual evolution of drug resistance, funding of ZIKV research once initiated will likely need to be sustained for several years, if not longer (e.g., HIV research has been funded for decades). As with other WCG projects, once scientists identify experimentally validated leads, finding a company to license them and pursue them in clinical trials and beyond will need incentives such as the FDA Tropical Disease Priority voucher, [9] which has a financial value on the open market [10].By working together and opening our research to the scientific community, many other labs will also be able to take promising molecular candidates forward to accelerate progress towards defeating the ZIKV outbreak. We invite any interested researcher to join us (send us your models or volunteer to assay the candidates we identify through this effort against any of the flaviviruses), and we hope new volunteers in the general public will donate their dormant, spare computing cycles to this cause. We will ultimately report the full computational and experimental results of this collaboration.

Advantages and Disadvantages of OpenZika

Advantages
  • Open Science could accelerate the discovery of new antivirals using docking and virtual screening
  • Docking narrows down compounds to test, which saves time and money
  • Free to use distributed computing on World Community Grid, and the workflow is simpler than using conventional supercomputers
Disadvantages
  • Concern around intellectual property ownership and whether companies will develop drugs coming from effort
  • Need for experimental assays will always be a factor
  • Testing in vitro and in vivo is not free, nor are the samples of the compounds
  相似文献   

2.
“Fit-for-purpose” diagnostic tests have emerged as a prerequisite to achieving global targets for the prevention, control, elimination, and eradication of neglected tropical diseases (NTDs), as highlighted by the World Health Organization’s (WHO) new roadmap. There is an urgent need for the development of new tools for those diseases for which no diagnostics currently exist and for improvement of existing diagnostics for the remaining diseases. Yet, efforts to achieve this, and other crosscutting ambitions, are fragmented, and the burden of these 20 debilitating diseases immense. Compounded by the Coronavirus Disease 2019 (COVID-19) pandemic, programmatic interruptions, systemic weaknesses, limited investment, and poor commercial viability undermine global efforts—with a lack of coordination between partners, leading to the duplication and potential waste of scant resources. Recognizing the pivotal role of diagnostic testing and the ambition of WHO, to move forward, we must create an ecosystem that prioritizes country-level action, collaboration, creativity, and commitment to new levels of visibility. Only then can we start to accelerate progress and make new gains that move the world closer to the end of NTDs.

Ahead of the second-ever World Neglected Tropical Disease (NTD) Day in January 2021, and amid the global Coronavirus Disease 2019 (COVID-19) crisis, the World Health Organization (WHO) launched a new roadmap for the prevention, control, elimination, and eradication of NTDs—a group of 20 diseases affecting more than one billion people worldwide [1]. Diagnostic testing is central to safeguarding decades of progress in NTDs and must be strategically leveraged to reach the goals laid out in the new NTD roadmap.Stepping back, we recognize the massive progress that has been made to combat NTDs. Today, 500 million fewer people need treatment for these debilitating diseases than in 2010, and 40 countries or areas have eliminated at least one of the 20 [1]. Yet, despite these gains, NTDs continue to impose a devastating human, social, and economic toll on the world’s poorest and most vulnerable communities [26]. COVID-19 is compounding the situation by wreaking havoc on health systems, which impacts progress on NTDs: this includes interruptions to mass treatment campaigns for diseases controlled through preventive chemotherapy (PCT) or individual case management interventions, as well as rerouting the already sparse available funding and resources [7].Diagnostic testing has been central to the COVID-19 response even with the introduction of vaccines. The rapid ramp up of research and development (R&D), the scaling up of low-cost and decentralized testing, and country-led approaches to tailored testing strategies for COVID-19, as well as lessons learned, can also provide new thinking around testing for NTDs. The new NTD roadmap offers a series of multisectoral actions and intensified, cross-cutting approaches to get us back on track—with diagnostics central to unlocking and accelerating this progress [1].However, the NTD roadmap shows that, of all 20 diseases or disease groups, just 2 (yaws and snakebite envenoming) are supported by adequate and accessible diagnostic tools. Six have no diagnostic tests available at all, with tools for each of the remaining conditions in urgent need of adaptation, modification, and/or improved accessibility (likely a more cost-effective option than the development of new diagnostics for these NTDs) [1]. This has to change. NTDs cannot continue to be neglected in favor of other competing priorities, or we risk losing the progress made to date.Until the COVID-19 pandemic thrust testing into the spotlight, diagnostics have been a “silent partner” in healthcare, receiving little by way of international attention and funding, specific country strategies, and dedicated budget lines. NTDs are no exception. Just 5% of the (limited) funding made available to NTDs has been invested in new diagnostics, compared with 44% and 39% on basic research and medicines and vaccines, respectively [1]. For most NTDs, diagnostics are a market failure situation, and as such, are not commercially viable enough to attract private investment. Consequently, very few diagnostic developers engage in this area—contrary, for example, to COVID-19, where developers are in the hundreds. Furthermore, as some diseases approach the last mile of elimination, falling infection rates precipitate the need for increasingly sensitive tests [1]. But progress in R&D is slow and fragmented, with a lack of engagement and coordination between governments, industry, donors, and development actors, leading to the duplication—and potential waste—of scant resources. While serial testing using multiple diagnostic tools or techniques can compensate for low sensitivity [8], such approaches are associated with increased costs of testing, sample collection, and transportation.Closing the diagnostic gap then, is a prerequisite to achieving the global ambition for NTDs, with the new NTD roadmap giving a blueprint for action. It is for this reason that we call on governments, industry, donors, and development actors to
  • Prioritize country-level diagnostic action: As we enter a new era in NTD management and control, we need to shift from traditional, donor-led models to country-driven initiatives. Government ministries must engage with, and advocate on behalf of, their poorest and most vulnerable populations so that no one is left behind. Political frameworks should prioritize diagnostics for NTDs in line with local disease burdens, and as part of fully funded, national health action plans that include a commitment to seeing the process through. Capacity building for diagnostics is also essential at country, sub-regional, and regional levels, including the establishment of laboratory networks, so that testing can be implemented in field settings.
  • Collaborate and create: There is never going to be a one-size-fits-all for NTD diagnostics. If targets are to be achieved, we need global frameworks that enable industry, manufacturers, and pharmaceutical companies to engage in the whole process, from R&D to supply chain logistics. Companies need to share knowledge, learnings, and innovation across multiple diseases. This will mean breaking silos and finding new ways to harness the power of existing products, technologies, and infrastructures. Further, it will mean creating economies of scale through regional manufacturing hubs and finding new, cross-cutting approaches to drive systemic change. To obtain the maximum access to technology and relevant intellectual property rights for NTD diagnostics, it is important to ensure that such rights are broadly available (non-exclusively) in NTD-endemic countries and are affordable (e.g., zero royalty rights).
  • Commit to new levels of visibility: The resources needed to realize that this ambition is limited, with a lack of visibility around the diagnostic landscape undermining progress in NTD management and control. Creating an ecosystem with visibility, transparency, and integration at its core will help streamline programmatic action, reduce the risk of duplication, and leverage the full potential from this limited pool. To do this, industry, donors, and other development actors must provide the information needed to map both funding and product landscapes. Using this information to create a virtual product pipeline will bring an unprecedented level of transparency to diagnostic developments—harmonizing multisectoral efforts and creating a robust information platform from which new collaborations, synergies, and innovation can grow. Developing an online open-access diagnostic pipeline for WHO NTD roadmap priority pathogens would serve multiple purposes: (i) drive advocacy to address critical product and funding gaps; and (ii) reduce the likelihood of duplication of efforts. Together, this would strengthen partnerships across all stakeholders, from donors to industry partners, to accelerate development, evaluation, and adoption of diagnostic solutions for NTDs. The newly established NTD Diagnostic Technical Advisory Group (DTAG) to WHO NTD department has already identified the priority diagnostic needs for NTD programs not only in terms of developing new tools, but also the accessibility of existing tools [9]. Several sub-groups that focus more narrowly on single diseases or specific topics (i.e., skin NTDs or cross-cutting) have been established and have been tasked to develop tool and biomarker agnostic target product profiles (TPPs), which are now available (for the most part) on WHO website for use by any diagnostic manufacturer to support development of their specific technology. Alignment with these diagnostic priorities by all stakeholders is strongly recommended to facilitate attainment of WHO 2030 NTD roadmap goals.
  • Establish NTD biobanks: Biobanks are required for the clinical evaluation and validation of new diagnostic tests. Establishing local biobanks would support a country-driven approach as well as allowing for head-to-head comparisons between tests and assessments of cross-reactivity across different NTDs.
  • Invest in existing diagnostics: The development of new diagnostics is a complex process, and the time from development to implementation can be lengthy. Training laboratory staff in the use of existing diagnostics and the establishment of robust quality control systems are effective approaches to achieving shorter-term improvements.
There is a long road ahead, but the past 10 years have shown us what can be achieved when governments, industry, donors, and development actors are bound by a shared, global goal. As we look forward to the next decade, we must prioritize country-level action, collaboration, creativity, and commitment to new levels of visibility, if we are to finally end the neglect of NTDs.  相似文献   

3.
Scabies has recently gained international attention, with the World Health Organization (WHO) recognizing it as a neglected tropical disease. The International Alliance for the Control of Scabies recently formed as a partnership of more than 15 different countries, with an aim to lead a consistent and collaborative approach to preventing and controlling scabies globally. Scabies is most prevalent in low-resource and low socioeconomic areas that experience overcrowding and has a particularly high prevalence in children, with an estimated 5% to 10% in endemic countries. Scabies is widespread in remote Aboriginal and Torres Strait Islander communities in Australia with the prevalence of scabies in Aboriginal and Torres Strait Islander children in remote communities estimated to be as high as 33%, making it the region with the third highest prevalence in the world. This population group also have very high rates of secondary complications of scabies such as impetigo, poststreptococcal glomerulonephritis (PSGN), and rheumatic heart disease (RHD). This article is a narrative review of scabies in remote Aboriginal and Torres Strait Islander populations in Australia, including clinical manifestations of disease and current treatment options and guidelines. We discuss traditional approaches to prevention and control as well as suggestions for future interventions including revising Australian treatment guidelines to widen the use of oral ivermectin in high-risk groups or as a first-line treatment.

Scabies has recently gained international attention, with the World Health Organization (WHO) recognizing it as a neglected tropical disease [1]. The International Alliance for the Control of Scabies recently formed as a partnership of more than 15 different countries, with an aim to lead a consistent and collaborative approach to preventing and controlling scabies globally [2]. In Australia, 10 million dollars was awarded to the Murdoch Children’s Research Institute to implement the World Scabies Elimination Program—an initiative aimed at collecting data from many affected countries and scaling up mass drug administration (MDA) [3].Scabies is most prevalent in low-resource and low socioeconomic areas that experience overcrowding and has a particularly high prevalence in children, with an estimated 5% to 10% in endemic countries [4,5]. The 2015 Global Burden of Disease Study ranked scabies with the 101st highest disability-adjusted life years (DALYs) estimate out of 246 conditions [6]. This is, however, likely underestimated as secondary complications, such as impetigo and kidney damage, were not included in this study [6,7]. A study from Fiji showed that 94% of impetigo was attributable to scabies [8], and it is estimated that approximately half of the instances of acute poststreptococcal glomerulonephritis (PSGN) in tropical regions can be attributed to skin infections [9].  相似文献   

4.
Two articles published earlier this year in the International Journal of Epidemiology [1,2] have re-ignited the debate over the World Health Organization’s long-held recommendation of mass-treatment of intestinal helminths in endemic areas. In this note, we discuss the content and relevance of these articles to the policy debate, and review the broader research literature on the educational and economic impacts of deworming. We conclude that existing evidence still indicates that mass deworming is a cost-effective health investment for governments in low-income countries where worm infections are widespread.  相似文献   

5.
Ribavirin is the only available Lassa fever treatment. The rationale for using ribavirin is based on one clinical study conducted in the early 1980s. However, reanalysis of previous unpublished data reveals that ribavirin may actually be harmful in some Lassa fever patients. An urgent reevaluation of ribavirin is therefore needed.

Fifty years after its discovery, Lassa fever remains uncontrolled, and mortality remains unacceptably high. Since 2015, Nigeria has been experiencing increasingly large outbreaks of Lassa fever, with new peaks reached in 2016, 2017, and 2018. In 1987, McCormick and colleagues reported a case fatality rate (CFR) of 16.5% among 441 patients hospitalized in Sierra Leone [1]. In Nigeria in 2019, 124 deaths were recorded among 554 laboratory-confirmed cases for a CFR of 22% [2].Ribavirin is the only available Lassa fever–specific treatment and has been used routinely for over 25 years. However, intravenous ribavirin is not licensed for Lassa fever. Its mechanism of action is unclear, it is expensive and hard to source, and it has well-known toxicities [3]. Therefore, the evidence for using ribavirin in Lassa fever deserves careful scrutiny. The emergence of potential new therapeutics for Lassa fever, such as favipiravir and monoclonal antibodies, adds further weight to the case for reconsidering the role of ribavirin since the evaluation of new drugs in clinical trials requires a comparison against existing treatments with a known efficacy and safety profile [4,5].The rationale for using ribavirin in Lassa fever is primarily based on one clinical study conducted in Sierra Leone in the late 1970s and early 1980s. McCormick and colleagues [6] reported that in Lassa fever patients with a serum aspartate aminotransferase (AST) level of ≥150 IU/L, the use of intravenous ribavirin within the first 6 days of illness reduced the fatality rate from 61% (11/18) with no ribavirin to 5% (1/20) (p = 0.002). These authors concluded that ribavirin is effective in the treatment of Lassa fever. However, there are long-standing concerns about the methods used in this study. Although randomization was used to assign patients to treatment groups, the comparisons presented were not according to original randomized groups, and we have reconstructed their derivation (Fig 1). Serious limitations to the comparisons presented include the use of historic controls, inclusion of pregnant women in the control group but their exclusion from the ribavirin group (case fatality is around 2-fold higher in pregnant women than nonpregnant patients), and post hoc merging of treatment groups. Despite this and the fact that the results only supported the use of ribavirin in nonpregnant adult patients with AST ≥150 IU/L, this study is the basis upon which ribavirin is now used in all patients with Lassa fever, including children, pregnant women, and people with normal liver function.Open in a separate windowFig 1Reconstruction of the McCormick et al. data.AST, aspartate aminotransferase; PW, pregnant women. † Discrepancy within McCormick et al, with 39 patients reported treated with oral ribavirin but only 38 (14+24) outcomes reported. ‡ Discrepancy within McCormick et al, with table 1 reporting 12/63 but text reporting 13/62.It has been well known among Lassa specialists that the McCormick study reports a subset of a much larger dataset assembled by the Lassa treatment unit in Sierra Leone and that a report on the full dataset was commissioned by the United States Army Medical Research and Development Command. One of us (PH) therefore submitted a freedom of information (FOI) request to access this report. The full report and an accompanying memo are available, and we encourage readers to access and read the materials [7,8]. The memo states that some of the original trial records were unavailable, and the data should be “interpreted with extreme caution.” Nonetheless, the report presents data from 1977 through to 1991 on 807 Lassa fever patients with a known outcome that were assigned to different ribavirin treatment regimens. These newly available data raise important questions about the safety and efficacy of ribavirin for the treatment of Lassa fever.The original data were lost during the civil war in Sierra Leone, but the report contains tables showing the distribution of characteristics of the whole population according to treatment group, an appendix showing individual data for the 405 patients who died, and results of a logistic regression analysis comparing the effect of ribavirin with no treatment for some of the ribavirin regimens, after adjusting for patient characteristics. Based on these data, we derived aggregated datasets containing the number of deaths according to treatment groups and individual characteristics. We combined groups I (“No treatment given”) and X (“Drugs were not available”) as no treatment and all groups in which ribavirin was administered (II, III, and V to IX) as ribavirin. Exhibit III-8 in the FOI report presented case fatality by treatment group and AST, from which we derived crude odds ratios (ORs) comparing ribavirin with no treatment. The logistic regression reported in Exhibit III-9 was restricted to “those treatment groups that yielded the lowest case fatality rates with respect to untreated patients in the high severity patient illness category” (groups II, III, V, and VII). It was adjusted for age, gender, time to admission, time to treatment, length of stay, and log(AST). We also reconstructed analyses by digitizing the data on individuals who died in Appendix D, calculating the number of deaths according to treatment group and AST, and subtracting these numbers from the totals presented in Exhibit III-2. These allowed us to estimate overall mortality ORs before and after adjusting for ribavirin, although the numbers did not entirely match, and so the number of deaths was reduced in some small groups.Estimates of the effect of oral and intravenous ribavirin from the McCormick study and of all ribavirin from the full report are shown in Fig 2. Based on the crude ORs derived from Exhibit III-8, ribavirin reduced mortality only in patients with serum AST ≥150 IU/L, with less benefit (OR 0.48 [95% CI 0.30 to 0.78]) than reported by McCormick and colleagues. However, ribavirin appeared to increase mortality in patients with serum AST <150 IU/L (2.90 [1.42 to 5.95]). In fact, in our analysis, the only stratum in which ribavirin appeared protective (0.38 [0.21 to 0.70]) was serum AST >300 IU/L (Table H in S1 Text). The logistic regression reported in the FOI report suggested a modest reduction in mortality, but the reasons for the choice of treatment groups compared were unclear. In the reconstructed analyses, ribavirin was associated with overall increased mortality (2.12 [1.67, 2.68]), although this was attenuated after adjustment for AST (1.48 [1.05, 2.08]).Open in a separate windowFig 2Forest plot of the OR of death in treatment and risk subgroups.AST, aspartate aminotransferase; FOI, freedom of information; OR, odds ratio.In our view, there is a compelling case to reevaluate the role of ribavirin in the care of patients with Lassa fever. The data suggest that ribavirin treatment may harm Lassa fever patients with AST <150 IU/L. The limitations revealed by the US Army report, such as large amounts of missing data, unclear treatment allocation practices, imbalances in treatment groups, and errors in coding serology results, cast further doubt on the conclusions of the McCormick study. This aligns with 2 recent systematic reviews by Eberhardt and colleagues and Cheng and colleagues, which concluded that the efficacy of ribavirin in Lassa fever was uncertain because of critical risk of bias in existing studies [9,10].Challenging a quarter of century of clinical practice is difficult. The first step is to acknowledge inadequacies in our knowledge and to ensure that treatment recommendations for Lassa fever better reflect the (weak) strength of evidence for ribavirin in different patient populations. Vigorous efforts should be made to engage clinicians and patients in designing a placebo-controlled trial to assess the safety and efficacy of ribavirin treatment in Lassa fever patients, particularly in those with milder disease (as may be indicated by an admission AST <150 IU/L) in whom the available evidence is compatible with ribavirin causing more harm than good.In conclusion, Lassa fever patients are receiving a drug that may lack efficacy or cause harm. It is incumbent on us to ensure that the next 25 years of Lassa fever treatment are built on more solid foundations.  相似文献   

6.
We describe an unusual case of type 2 leprosy reaction (T2R) with septic shock–like features induced by helminth infection in a 31-year-old Moluccan male patient with a history of completed treatment of WHO multidrug therapy (MDT)–multibacillary (MB) regimen 2 years before admission. During the course of illness, the patient had numerous complications, including septic shock, anemia, and disseminated intravascular coagulation (DIC). Nevertheless, antibiotic therapies failed to give significant results, and the source of infection could not be identified. Helminth infection was subsequently revealed by endoscopic examination followed by parasitological culture. Resolution of symptoms and normal level of organ function–specific markers were resolved within 3 days following anthelmintic treatment. This report demonstrated the challenge in the diagnosis and treatment of severe T2R. Given that helminth infections may trigger severe T2R that mimics septic shock, health professionals need to be aware of this clinical presentation, especially in endemic regions of both diseases.

Type 2 leprosy reaction (T2R) is a type III hypersensitivity reaction that can occur in people with lepromatous or borderline lepromatous leprosy before, during, or after completion of multidrug therapy (MDT). Its clinical manifestations are highly variable, which can be limited to the skin or accompanied by systemic disruption [1,2]. Uncommonly, it may also present with fever, hypotension, and tachycardia that mimic septic shock [3]. Helminth infections have been demonstrated to modulate the host immune response and induce leprosy reaction [4]. While concurrent helminth infections may benefit true sepsis by preventing exaggerated inflammation and severe pathology [5], treating helminth coinfection contributed directly to the dramatic improvement of the patient’s clinical and laboratory outcomes in this report.  相似文献   

7.

Background

Rabies is a fatal encephalitis caused by viruses belonging to the genus Lyssavirus of the family Rhabdoviridae. It is a viral disease primarily affecting mammals, though all warm blooded animals are susceptible. Experimental rabies virus infection in birds has been reported, but naturally occurring infection of birds has been documented very rarely.

Principal Findings

The carcass of a domestic fowl (Gallus domesticus), which had been bitten by a stray dog one month back, was brought to the rabies diagnostic laboratory. A necropsy was performed and the brain tissue obtained was subjected to laboratory tests for rabies. The brain tissue was positive for rabies viral antigens by fluorescent antibody test (FAT) confirming a diagnosis of rabies. Phylogenetic analysis based on nucleoprotein gene sequencing revealed that the rabies virus strain from the domestic fowl belonged to a distinct and relatively rare Indian subcontinent lineage.

Significance

This case of naturally acquired rabies infection in a bird species, Gallus domesticus, being reported for the first time in India, was identified from an area which has a significant stray dog population and is highly endemic for canine rabies. It indicates that spill over of infection even to an unusual host is possible in highly endemic areas. Lack of any clinical signs, and fewer opportunities for diagnostic laboratory testing of suspected rabies in birds, may be the reason for disease in these species being undiagnosed and probably under-reported. Butchering and handling of rabies virus- infected poultry may pose a potential exposure risk.  相似文献   

8.
Several issues have been identified with the current programs for the elimination of onchocerciasis that target only transmission by using mass drug administration (MDA) of the drug ivermectin. Alternative and/or complementary treatment regimens as part of a more comprehensive strategy to eliminate onchocerciasis are needed. We posit that the addition of “prophylactic” drugs or therapeutic drugs that can be utilized in a prophylactic strategy to the toolbox of present microfilaricidal drugs and/or future macrofilaricidal treatment regimens will not only improve the chances of meeting the elimination goals but may hasten the time to elimination and also will support achieving a sustained elimination of onchocerciasis. These “prophylactic” drugs will target the infective third- (L3) and fourth-stage (L4) larvae of Onchocerca volvulus and consequently prevent the establishment of new infections not only in uninfected individuals but also in already infected individuals and thus reduce the overall adult worm burden and transmission. Importantly, an effective prophylactic treatment regimen can utilize drugs that are already part of the onchocerciasis elimination program (ivermectin), those being considered for MDA (moxidectin), and/or the potential macrofilaricidal drugs (oxfendazole and emodepside) currently under clinical development. Prophylaxis of onchocerciasis is not a new concept. We present new data showing that these drugs can inhibit L3 molting and/or inhibit motility of L4 at IC50 and IC90 that are covered by the concentration of these drugs in plasma based on the corresponding pharmacological profiles obtained in human clinical trials when these drugs were tested using various doses for the therapeutic treatments of various helminth infections.

Onchocerca volvulus is an obligate human parasite and the causative agent for onchocerciasis, which is a chronic neglected tropical disease prevalent mostly in the sub-Saharan Africa. In 2017, 20.9 million people were infected, with 14.6 million having skin pathologies and 1.15 million having vision loss [1]. The socioeconomic impact of onchocerciasis and the debilitating morbidity caused by the disease prompted the World Health Organization (WHO) to initiate control programs that were first focused on reducing onchocerciasis as a public health problem, and since 2012, the ultimate goal is to eliminate it by 2030 [2]. Over the years, WHO sponsored and coordinated 3 major programs: The Onchocerciasis Control Programme (OCP), the African Programme for Onchocerciasis Control (APOC), and the Onchocerciasis Elimination Program of the Americas (OEPA). Since 1989, the control measures depended on mass drug administration (MDA) annually or biannually with ivermectin, which targets the transmitting stage of parasite, the microfilariae [35]. However, several issues have been identified with the current MDA programs including the need to expand the treatment to more populations depending on baseline endemicity and transmission rates [2,6]. Moreover, it became apparent that alternative and/or complementary treatment regimens as part of a more comprehensive strategy to eliminate onchocerciasis are needed [2]. Ivermectin has only mild to moderate effects on the adult stages of the parasite [79], and there are communities in Africa where the effects of ivermectin are suboptimal [10]. It is also contraindicated in areas of Loa loa co-endemicity [11], as well as in children under the age of 5 and in pregnant women. By relying only on MDA with ivermectin, the most optimistic mathematical modeling predicts that elimination will occur only in 2045 [12].To support the elimination agenda, much of the recent focus has been on improving efficacy outcomes through improved microfilariae control with moxidectin and the discovery of macrofilaricidal drugs that target the adult O. volvulus parasites [1318]. We posit that the addition of “prophylactic” drugs or therapeutic drugs that can be utilized in a prophylactic strategy to the toolbox of present microfilaricidal drugs and/or future macrofilaricidal treatment regimens will not only improve the chances of meeting the elimination goals but may also hasten the time for elimination and support achieving a sustained elimination of onchocerciasis. These “prophylactic” drugs will target the infective third- (L3) and fourth-stage (L4) larvae of O. volvulus and consequently prevent the establishment of new infections not only in the uninfected individuals but also in the already infected individuals and thus reduce the overall adult worm burden and transmission. Importantly, an effective prophylactic treatment regimen can utilize drugs that are already part of the onchocerciasis elimination program (ivermectin), those being considered for MDA (moxidectin) [19,20], and/or the potential macrofilaricidal drugs (oxfendazole and emodepside) currently under clinical development [21].Prophylaxis of onchocerciasis is not a new concept. In the 1980s, once ivermectin was introduced as a “prophylactic” drug against the filarial dog heartworm, Dirofilaria immitis [22], its prophylactic effects were also examined in Onchocerca spp. In chimpanzees, a single dose of ivermectin (200 μg/kg) was highly protective (83% reduction in patent infections) when given at the time of the experimental infection and tracked for development of patency over 30 months. It was, however, much less effective (33% reduction in patent infections) when given 1 month postinfection with the L3s, at which time the L4s had already developed [23]. Moreover, monthly treatment with ivermectin at either 200 μg/kg or 500 μg/kg for 21 months completely protected naïve calves against the development of O. ochengi infection as compared to untreated controls, which were 83% positive for nodules and 100% positive for patency [24]. When naïve calves exposed to natural infection were treated with either ivermectin (150 μg/kg) or with moxidectin (200 μg/kg) monthly or quarterly, none of the animals developed detectable infections after 22 months of exposure, except 2 animals in the quarterly ivermectin treated group which had 1 nodule each; in the non-treated control group, the nodule prevalence was 78.6% [25]. These prophylactic studies in calves exposed to natural infections clearly demonstrated that monthly or quarterly treatments with ivermectin and/or moxidectin over 22 months were highly efficacious against the development of new infections. When ivermectin was administered in a highly endemic region of onchocerciasis in Cameroon every 3 months over a 4-year period, it resulted in reduced numbers of new nodules (17.7%) when compared to individuals who were treated annually. This recent study suggests that ivermectin may have also a better prophylactic effect in humans when administered quarterly [26].Importantly, moxidectin, a member of the macrocyclic lactone family of anthelmintic drugs, also used in veterinary medicine like ivermectin [20], was recently approved for the treatment of onchocerciasis as a microfilaricidal drug in individuals over the age of 12 [20]. In humans, a single dose of moxidectin (8 mg) appeared to be more efficacious than a single dose of ivermectin (150 μg/kg) in terms of lowering microfilarial loads [17]. Modeling has shown that an annual treatment with moxidectin and a biannual treatment with ivermectin would achieve similar reductions in the duration of the MDA programs when compared to an annual treatment with ivermectin [27].In our efforts to identify macrofilaricidal drugs, we tested a selection of drugs for their ability to inhibit the molting of O. volvulus L3 to L4 as part of the in vitro drug screening funnel [13,2831]. With some being highly effective, we decided to also examine the effects of the known MDA drugs and those already in clinical development for macrofilaricidal effects on molting of L3 and the motility of L4 (S1 Text) as potential “prophylactic” drugs. When ivermectin and moxidectin were evaluated, we found that both drugs were highly effective as inhibitors of molting: IC50 of 1.048 μM [918.86 ng/ml] and IC90 of 3.73 μM [2,949.1 ng/ml] for ivermectin and IC50 of 0.654 μM [418.43 ng/ml] and IC90 of 1.535 μM [985.3 ng/ml] for moxidectin (Table 1 and S1 Fig), with moxidectin being more effective than ivermectin. When both drugs were tested against the L4, we found that both drugs inhibited the motility of L4s after 6 days of treatment: Ivermectin had an IC50 of 1.38 μM [1,207.6 ng/ml] and IC90 of 31.45 μM [27,521.9 ng/ml] (Table 1 and S1 Fig), while moxidectin had an IC50 of 1.039 μM [665.4 ng/ml] and IC90 of approximately 30 μM [approximately 19,194 ng/ml] (Table 1 and S1 Fig). Interestingly, when the treatment of L4 with both drugs was prolonged, the IC50 values for the inhibition of L4 motility on day 11 with ivermectin and moxidectin were 0.444 μM and 0.380 μM, respectively. Significantly, from the prospect of employing both drugs for prophylaxis against new infections with O. volvulus, moxidectin (8 mg) has an advantage as it achieves a maximum plasma concentration of 77.2 ± 17.8 ng/ml, is metabolized minimally, and has a half-life time of 40.9 ± 18.25 days with an area under the curve (AUC) of 4,717 ± 1,494 ng*h/ml in healthy individuals [32], which covers the experimental IC50 achieved by moxidectin for inhibiting both L3 molting and L4 motility, and the IC90 for L3s. In comparison, ivermectin reaches a maximum plasma concentration of 54.4 ± 12.2 ng/ml with a half-life of 1.5 ± 0.43 days and an AUC of 3,180 ± 1,390 ng*h/ml in healthy humans [33], which only covers the IC50 for inhibiting molting of L3 and motility of L4. We therefore reason that based on the significantly improved pharmacokinetic profile of moxidectin and its efficacy against both L3 and L4 larvae in vitro (Table 1), it might have a better “prophylactic” profile than ivermectin for its potential to interrupt the development of new O. volvulus infections, and thus ultimately affect transmission and further support the elimination of onchocerciasis. Adding to moxidectin’s significance, in dogs, it is a highly effective prophylactic drug against ivermectin-resistant D. immitis strains [19], an important attribute in the event that a suboptimal responsiveness to ivermectin treatment becomes more widespread in the onchocerciasis endemic regions of Africa. Testing the potential effect of moxidectin on the viability or development of transmitted L3 larvae was already recommended by Awadzi and colleagues in 2014 [34], when the excellent half-life of moxidectin in patients with onchocerciasis was realized. We have to acknowledge, however, that the key parameters that can predict the potency of a drug is actually a combination of exposure (drug concentrations) at the site of action and the duration of that exposure that is above the determined IC50/IC90. As we have access to only the AUC, half-life, and Cmax data for each of the in vitro–tested drugs, the use of plasma concentrations for predicting the anticipated potency of these putative “prophylactic” drugs in vivo has to be further assessed with care during clinical trials.Table 1Inhibition of O. volvulus L3 molting and L4 motility in vitro by the prospective prophylactic drugs and their essential pharmacokinetic parameters at doses currently used or deemed safe for use in humans.
DrugIvermectinMoxidectinAlbendazoleOxfendazoleEmodepside
Albendazole sulfoxide
IC50 μM
(conc in ng/ml)
IC90 μM
(conc in ng/ml)
IC50 μM
(conc in ng/ml)
IC90 μM
(conc in ng/ml)
IC50 μM
(conc in ng/ml)
IC90 μM
(conc in ng/ml)
IC50 μM
(conc in ng/ml)
IC90 μM
(conc in ng/ml)
IC50 μM
(conc in ng/ml)
IC90 μM
(conc in ng/ml)
In vitro drug testing with O. volvulus larvaeInhibition of L3 moltinga1.048 (918.86 ng/ml)3.730 (2,949.1 ng/ml)0.654 (418.43 ng/ml)1.535 (985.3 ng/ml)0.007 (1.9 ng/ml)0.023 (5.8 ng/ml)0.034 (10.7 ng/ml)0.071 (22.4 ng/ml)0.0007 (0.8 ng/ml)0.002 (2.2 ng/ml)
0.008 (2.25 ng/ml)0.07 (19.69 ng/ml)
Inhibition of L4 motilityb1.38 (1,207 ng/ml)31.45 (27,521 ng/ml)1.039 (665 ng/ml)approximately 30 (approximately 19,194 ng/ml)>2 μM0.0005 (0.6 ng/ml)0.078 (87.3 ng/ml)
Pharmacokinetic profiles extracted from data collected during clinical trials in humanscDose150 μg/kg8 mg400 mg15 mg/kg30 mg/kg1 mg40 mg
Cmax (plasma) ng/ml54.4 ± 12.277.2 ± 17.824.5288d6,250 ± 1,3905,300 ± 1,69018.6434
Half-life t1/2 (h)36.6 ± 10.2981 ± 4381.538.56d9.97 ± 2.229.82 ± 3.4642.7392
AUC (ng*h/ml)3,180 ± 1,3904,717 ± 1,494733,418d99,500 ± 2,44078,300 ± 2,8301003,320
Citations[33][32]e[41][42][43]
Open in a separate windowaO. volvulus L3 obtained from infected Simulium sp. were washed and distributed at n = approximately 10 larvae per well and cocultured in contact with naïve human peripheral blood mononuclear cells for a period of 6 days with or without the respective drugs in vitro (S1 Text) and as previously described [13,30]. Ivermectin (PHR1380, Sigma-Aldrich, St. Louis, Missouri, United States of America) and moxidectin (PHR1827, Sigma-Aldrich) were tested in the range of 0.01–10 μM; albendazole (A4673, Sigma-Aldrich), albendazole sulfoxide (35395, Sigma-Aldrich), and oxfendazole (31476, Sigma-Aldrich) in the range of 1–3 μM; and emodepside (Bayer) in the range of 0.3–1 μM using 3-fold dilutions. On day 6, molting of L3 worms was recorded. Each condition was tested in duplicate and repeated at least once. The IC50 and IC90 were derived from nonlinear regression (curve fit) analysis on GraphPad Prism 6 with 95% confidence intervals.bL3s were allowed to molt to L4 in the presence of PBMCs and on day 6 when molting was complete the L4 larvae were collected and distributed at 6–8 worms per well and treated with the respective concentrations of drugs [ivermectin and moxidectin: 0.01–30 μM at 3-fold dilutions and emodepside: 0.03–3 μM at 10-fold dilutions and 10 μM] for a period of 6 days. Inhibition of O. volvulus L4 motility was recorded as described [13,30]; representative videos of motility and inhibited motility can be viewed in Voronin and colleagues [30], S1–S3 Videos. Each condition was tested in duplicate and repeated at least once. The IC50 and IC90 were derived from nonlinear regression (curve fit) analysis on GraphPad Prism 6 with 95% confidence intervals.cInformation regarding the pharmacokinetic profiles of each drug was extracted from public data collected during the corresponding clinical trial(s) in humans, which are also referenced.dPharmacokinetic parameters of albendazole sulfoxide, the predominant metabolite of albendazole.eAdditional pharmacokinetics parameters for moxidectin not only in heathy individual but also in those living in Africa can be found on the moxidectin FDA prescribing information website: https://www.drugs.com/pro/moxidectin.html. In patients with onchocerciasis, it is reported that a single dose of moxidectin (8 mg) achieves a maximum plasma concentration of 63.1 ± 20.0 ng/ml, and it has a half-life time of 559 ± 525 days with an AUC of 2,738 ± 1,606 ng*h/ml.AUC, area under the curve; Cmax, maximum plasma concentration.The prospects for identifying additional “prophylactic” drugs against O. volvulus increased when we tested 3 other drugs: albendazole, already in use for controlling helminth infections in humans; and oxfendazole and emodepside, being tested by the Drugs for Neglected Diseases initiative (DNDi) as potential repurposed macrofilaricidal drugs for human indications [21]. Albendazole is a primary drug of choice for MDA treatment of soil-transmitted helminths (STH; hookworms, whipworms [in combination with oxantel pamoate], and ascarids) [35], as well as for the elimination of lymphatic filariasis in Africa when used in combination with ivermectin [36]. Oxfendazole, a member of the benzimidazole family, is currently indicated for the treatment of a range of lung and gastrointestinal parasites in cattle and other veterinary parasites and is favorably considered for the treatment and control of helminth infections in humans [37]. Emodepside, an anthelmintic drug of the cyclooctadepsipeptide class, is used in combination with praziquantel to treat a range of gastrointestinal nematodes in dogs and cats [3840].We found that all 3 drugs were highly effective at inhibiting the molting of O. volvulus, even more than ivermectin or moxidectin. The IC50 for inhibition of L3 molting with albendazole was 7 nM [1.9 ng/ml], and the IC90 was 23 nM [5.8 ng/ml]. The IC50 for inhibition of L3 molting with oxfendazole was 34 nM [10.7 ng/ml], and the IC90 was 71 nM [22.4 ng/ml] (Table 1 and S1 Fig). Albendazole and oxfendazole were less effective at inhibiting the motility of L4s, both having IC50 >2 μM (Table 1). In previous studies, we reported that tubulin-binding drugs (flubendazole and oxfendazole) affected the motility of L4s and L5s only after repeated treatments over 14 days in culture [13,30]. Hence, both drugs might be more effective against L3s than L4s, a stage that may require prolonged treatments and further evaluation with future studies. Albendazole is used for STH treatment as a single dose of 400 mg. At this dose, it reaches a maximum plasma concentration of 24.5 ng/ml with a half-life time of 1.53 hours (AUC of 73 ng*h/ml) [41], which covers the IC90 for inhibition of L3 molting. In comparison, albendazole sulfoxide, an important active metabolite of albendazole, had a much improved maximum plasma concentration of 288 ng/ml with a half-life time of 8.56 hours (AUC of 3,418 ng*h/ml) than albendazole [41] (Table 1), and which covers the IC50 of 8 nM [2.25 ng/ml] and IC90 of 70 nM [19.69 ng/ml] for inhibition of L3 molting in vitro. Oxfendazole, when administered at the doses currently being tested for efficacy against trichuriasis (whipworm infection), 30 mg/kg and 15 mg/kg, achieved a maximum plasma concentration of 5,300 ± 1,690 and 6,250 ± 1,390 ng/ml, respectively, with a half-life time of approximately 9.9 hours (AUC: 78,300 ± 2,830 to 99,500 ± 2,440 ng*h/ml) (Table 1) [42], both of which cover the IC90 for inhibition of L3 molting. Hence, from the perspective of preventing newly established infections with O. volvulus L3 by inhibiting their molting, oxfendazole and albendazole are additional compelling candidates to consider.Intriguingly, emodepside was the most effective drug on both L3s and L4s; it inhibited molting with an IC50 of 0.7 nM [0.8 ng/ml] (which is 10, 48.5, and approximately 1,000 times more potent than albendazole, oxfendazole, and moxidectin, respectively) and an IC90 of 2 nM [2.2 ng/ml]. Importantly, it also inhibited the motility of L4s by day 6 with an IC50 of 0.5 nM [0.6 ng/ml] and an IC90 of 78 nM [87.3 ng/ml] (Table 1 and S1 Fig), which is also more potent than the other drugs. In the ascending dose (1 to 40 mg) human clinical trial (NCT02661178), emodepside achieved a maximum plasma concentration in the range of 18.6 to 595 ng/ml, AUC of 100 to 4,112 ng*h/ml, and half-life of 1.7 to 24.6 days depending on the dose administered, and all doses were well-tolerated (Table 1) [43]. Considering that the IC90 for inhibition of L3 molting and L4 motility in vitro are 2 nM and 78 nM (Table 1 and S1 Fig), respectively, these values are already covered by the PK profile of the drug starting at 2.5 mg. Hence, the clinical trials for emodepside as a macrofilaricidal drug, if efficacious at 2.5 mg or above, could have additional implications in terms of utilizing emodepside for prophylactic potential.We propose that all 5 drugs are effective against the early stages of O. volvulus based on their efficacy (IC50/IC90) in vitro. However, based on their known pharmacokinetic profiles in humans, they can be prioritized for future evaluation for their utility for prophylactic activity in humans as follows: emodepside > moxidectin > albendazole > oxfendazole > ivermectin. Moreover, we believe that the addition of some of these putative “prophylactic” drugs individually or in combination with the current MDA regimens against onchocerciasis would also align well with the integrated goals of the Expanded Special Project for Elimination of Neglected Tropical Diseases and possibly also expedite the elimination goals of one of the other 6 neglected tropical diseases amenable to MDA: the STH [44]. All 5 of these drugs are broad-spectrum anthelmintic drugs that are effective against STH infections [4549], and thus may also benefit MDA programs aimed at controlling STH infections. The effects of MDA with ivermectin or albendazole on STHs (hookworms, Ascaris lumbricoides, and Trichuris trichiura) have already been explored in clinical studies [45,47,50] and were shown to have a significant impact on the STH infection rates in the treated communities. One dose of moxidectin (8 mg) in combination with albendazole (400 mg) was as effective as a combination of albendazole and oxantel pamoate (currently the most efficacious treatment against T. trichiura) in reducing fecal T. trichiura egg counts [46]. Notably, oxfendazole is also being tested for its effectiveness in humans against trichuriasis (NCT03435718). Additionally, emodepside was shown to not only have a strong inhibitory activity against adult STH worms in animal models with an ED50 of less than 1.5 mg/kg, but also against STH larval stages in vitro with IC50 <4 μM for L3s [49].We could envision that a single drug, a combination of any of these 5 drugs, or just those we have prioritized (moxidectin and emodepside), when administered also for prophylaxis against the development of new O. volvulus infection, would also protect against new STH infections. Broad-spectrum chemoprophylaxis of nematode infections in humans could potentially also save on costs and time invested toward elimination of co-endemic parasites through the administration of a combination of drugs. Moreover, considering the time-consuming process of drug discovery, the heavy costs incurred, and the excessive failure rates, the prospect of repurposing commercially available drugs used for other human or veterinary diseases for the prophylaxis of O. volvulus infection is an attractive one [31,5154]. Repurposing of drugs could also accelerate the approval timeline for new drug indications since information regarding mechanism, dosing, toxicity, and metabolism would be readily available.In summary, our O. volvulus in vitro drug testing studies reinforce the “old” proposition of employing MDA drugs for prophylactic strategies as well, inhibiting the development of new infections with O. volvulus in the endemic regions under MDA. We report for the first time that in vitro, emodepside, moxidectin, and ivermectin have very promising inhibitory effect on both L3s and L4s, with albendazole and oxfendazole for additional consideration. Importantly, considering that the L4 larvae are longer lived as compared to the L3 stage, and hence the more feasible target against the establishment of new infections, we believe that targeting the L4 stage would be an invaluable tool toward advancing sustainable elimination goals for onchocerciasis. Moxidectin and emodepside with their superior half-life and pharmacokinetic profiles in humans and their efficacy in vitro against both L3 and L4 stages of the parasite seem to show the most promise for this purpose. Of significance, the doses required to provide exposures that would cover the IC90 achieved by these 2 drugs in vitro against L3 and emodepside against L4 have been shown to be well-tolerated in humans (Table 1). Crucially, as these new drugs are rolled out for human use as microfilaricidal and/or macrofilaricidal drugs, it would be important to add to the clinical protocols to also observe their effects on the development of new infections in populations that are exposed to active transmission using serological assays that can predict new infections and distinguish them from earlier infections [55]. This could potentially reveal valuable information to foster the development of more complementary elimination programs that not only target the microfilariae (moxidectin) and the adult worms (emodepside) but also the other infectious stages of the parasite, with their effects on STH being an added advantage.Mathematical modeling has long influenced the design of intervention policies for onchocerciasis and predicted the potential outcomes of various regimens used by the elimination programs and the feasibility of elimination [5660]. We believe that a revised mathematical model that also takes into account the additional aspect of targeting L3 and L4 stages could be helpful to assess the enhanced impact this complementary tool might have in advancing the goal of elimination, and accordingly support a revised policy for operational intervention programs first for onchocerciasis, and perhaps also as a pan-nematode control measure, by the decision-making bodies [7,61,62]. Given that in human clinical trials in which infected people were treated quarterly with ivermectin, there was an indication of a considerable trend of reduced number of newly formed nodules, it becomes apparent that the recommendation for such a revised regimen might also support protection from new infections. Clinical trials to assess the efficacy of biannual doses of ivermectin or moxidectin versus annual doses of these drugs against onchocerciasis have been already initiated (NCT03876262). Alternatively, increasing the frequency of future treatments with moxidectin and/or emodepside to biannual or quarterly treatment and/or using them in combinations could also improve their chemotherapeutic potential by targeting multiple stages of the parasite, thus increasing all the control potential of these new MDA drugs on multiple stages of the parasite and ultimately support not only a faster timeline but also sustained elimination.  相似文献   

9.
Rabies diagnosis proficiency tests on animal specimens using four techniques (FAT, RTCIT, conventional RT-PCR and real-time RT-PCR) were organised over 10 years (2009–2019). Seventy-three laboratories, of which 59% were from Europe, took part. As the panels were prepared with experimentally-infected samples, the error rate of laboratories on positive and negative samples was accurately estimated. Based on fitted values produced by mixed modelling including the variable “laboratory” as a random variable to take into account the longitudinal design of our dataset, the technique that provided the most concordant results was conventional RT-PCR (99.3%; 95% CI 99.0–99.6), closely followed by FAT (99.1%; 95% CI 98.7–99.4), real-time RT-PCR (98.7%; 95% CI 98.1–99.3) and then RTCIT (96.8%; 95% CI 95.8–97.7). We also found that conventional RT-PCR provided a better diagnostic sensitivity level (99.3% ±4.4%) than FAT (98.7% ±1.6%), real-time RT-PCR (97.9% ±0.8%) and RTCIT (95.3% ±5.1%). Regarding diagnostic specificity, RTCIT was the most specific technique (96.4% ±3.9%) followed closely by FAT (95.6% ±3.8%), real-time RT-PCR (95.0% ±1.8%) and conventional RT-PCR (92.9% ±0.5%). Due to multiple testing of the samples with different techniques, the overall diagnostic conclusion was also evaluated, and found to reach an inter-laboratory concordance level of 99.3%. The concordance for diagnostic sensitivity was 99.6% ±2.0% and for diagnostic specificity, 98.0% ±8.5%. Molecular biology techniques were, however, found to be less specific than expected. The potential reasons for such findings are discussed herein. The regular organisation of performance tests has contributed to an increase in the performance of participating laboratories over time, demonstrating the benefits of such testing. Maintaining a high-quality rabies diagnosis capability on a global scale is key to achieving the goal of eliminating dog-mediated human rabies deaths. The regular organisation of exercises on each continent using selected local strains to be tested according to the local epidemiological situation is one factor that could help increase reliable diagnosis worldwide. Rabies diagnosis capabilities could indeed be enhanced by providing adequate and sustainable proficiency testing on a large scale and in the long term  相似文献   

10.

Background

There are few detailed etiologic studies of severe anemia in children from malaria-endemic areas and none in those countries with holoendemic transmission of multiple Plasmodium species.

Methodology/Principal Findings

We examined associates of severe anemia in 143 well-characterized Papua New Guinean (PNG) children aged 0.5–10 years with hemoglobin concentration <50 g/L (median [inter-quartile range] 39 [33][44] g/L) and 120 matched healthy children (113 [107–119] g/L) in a case-control cross-sectional study. A range of socio-demographic, behavioural, anthropometric, clinical and laboratory (including genetic) variables were incorporated in multivariate models with severe anemia as dependent variable. Consistent with a likely trophic effect of chloroquine or amodiaquine on parvovirus B19 (B19V) replication, B19V PCR/IgM positivity had the highest odds ratio (95% confidence interval) of 75.8 (15.4–526), followed by P. falciparum infection (19.4 (6.7–62.6)), vitamin A deficiency (13.5 (5.4–37.7)), body mass index-for-age z-score <2.0 (8.4 (2.7–27.0)) and incomplete vaccination (2.94 (1.3–7.2)). P. vivax infection was inversely associated (0.12 (0.02–0.47), reflecting early acquisition of immunity and/or a lack of reticulocytes for parasite invasion. After imputation of missing data, iron deficiency was a weak positive predictor (6.4% of population attributable risk).

Conclusions/Significance

These data show that severe anemia is multifactorial in PNG children, strongly associated with under-nutrition and certain common infections, and potentially preventable through vitamin A supplementation and improved nutrition, completion of vaccination schedules, and intermittent preventive antimalarial treatment using non-chloroquine/amodiaquine-based regimens.  相似文献   

11.
Neurocysticercosis (NCC), the infection of the nervous system by the cystic larvae of Taenia solium, is a highly pleomorphic disease because of differences in the number and anatomical location of lesions, the viability of parasites, and the severity of the host immune response. Most patients with parenchymal brain NCC present with few lesions and a relatively benign clinical course, but massive forms of parenchymal NCC can carry a poor prognosis if not well recognized and inappropriately managed. We present the main presentations of massive parenchymal NCC and their differential characteristics.

Infection of the central nervous system by the larval stage of Taenia solium—the pork tapeworm—causes neurocysticercosis (NCC), a highly pleomorphic disease [1]. This pleomorphism is partly related to differences in the number and anatomical location of lesions, the viability of parasites, and the severity of the host immune response against the infection. Cysticerci may be located within the brain parenchyma, the subarachnoid space, the ventricular system, the spinal cord, the sellar region, or even the subdural space.Most patients with parenchymal NCC present with few lesions and a clinical course that is often more benign than that observed in the subarachnoid and ventricular forms of NCC, where a sizable proportion of patients are left with disabling sequelae or may even die as a result of the disease [2,3]. Nevertheless, massive forms of parenchymal NCC require special attention to reduce the risk of complications related to the disease itself or to an inadequate treatment. Here, we present the main presentations of massive parenchymal NCC and their differential characteristics. There is no standardized definition of how many cysts constitute massive NCC. While the term “massive” has usually been applied when there are more than 100 lesions in the brain parenchyma, others have used smaller numbers (50), and there is not a defined cutoff.  相似文献   

12.
Interest in filariasis has found a new impetus now that neglected tropical diseases have their own journal. However, some of the advances published in renowned international journals have completely ignored previous publications on the subject, particularly those in languages other than English. The rapid assessment procedure for loiasis and the mapping of lymphatic filariasis provide two perfect illustrations of this. This problem may seem a bit outdated, given that all “good authors” now publish exclusively in English. It certainly is outdated for most areas of medicine. But, surely, this should not be the case for neglected tropical diseases, for which certain long-standing findings are every bit as important as what may be presented as new discoveries. One possibility would be for certain journals, such as PLOS Neglected Tropical Diseases, to include a specific heading permitting the publication in English of older studies that initially appeared in a language other than English. The texts would be English versions respecting the entirety of the original text. Submission should be accompanied by a presentation of the problem, with details and explanatory comments, with submission at the initiative of the authors of the former article in question or their students or sympathizers.Interest in filariasis has found a new impetus now that neglected tropical diseases (NTDs) have their own journal. However, some of the advances published in renowned international journals have completely ignored previous publications on the subject, particularly those in languages other than English. This Viewpoint article is intended to make us ponder the issue of a language gap or discrimination existing in publishing outcomes and reference citations. This is also the question of deleterious effects of the obligation “to be in English or not to be”.The rapid assessment procedure for loiasis (RAPLOA) and the geographical distribution of lymphatic filariasis provide two perfect illustrations of this.The RAPLOA has recently been widely used to determine the regional endemicity of loiasis and to update existing endemicity data for this disease over its global distribution range. This important work has been recently published in PLOS NTDs [1]. The determination, within a population of the prevalence or, preferably, the annual incidence of episodes, of conjunctival migration by adult worms is a simple, non-invasive, relatively sensitive and specific method for evaluating the endemicity of Loa loa. This approach has proved particularly useful in areas in which both loiasis and onchocerciasis are observed: the mass treatment program to control onchocerciasis is based on the use of ivermectin and there is a risk of adverse treatment outcomes in patients carrying large numbers of L. loa worms [2]. In regions of high endemicity, the correlation between the conjunctival migration index and the microfilarial index is strong overall, both for villages and for age groups. Its use as an epidemiological index was clearly proposed in a publication in 1994 [3]. However, as this article was published in French, in Médecine Tropicale (Marseille), it has never been cited, despite being listed in international databases, including PubMed. A poster communication concerning the same issue had no real impact either, despite being presented at an international congress [4]. The origin of this new epidemiological index (RAPLOA) is systematically attributed to two World Health Organization (WHO) publications in 2001 [5] and 2002 [6]. It is true that the studies reported in these publications validated the concept at a large scale and in different endemic foci.The usefulness of specific clinical manifestations (eye worm and Calabar swelling) to assess L. loa had been recommended by different authors as early as 1950 [7]. But the correlation between the microfilarial index and the frequency of ocular migration has not been studied, and even less attention was paid to the notion of an epidemiological index until the epidemiological studies carried out in Congo Republic (former People''s Republic of the Congo) during the 1980s [8]. However, the index as such was clearly defined in 1994 [3]. Here is a direct translation of an excerpt of the French text published in 1994: “For loiasis, the usual parasitological indices (microfilarial index and mean microfilarial density) are the only measures recognized as providing information about the level of endemicity in humans. In addition to requiring blood samples standardized in terms of both volume and sampling time, these indices do not reflect the real level of parasitism, given the high frequency of infected subjects without microfilaria in the blood. Subjects infested with mature, fertile adult worms, as demonstrated by the removal of a subconjunctival female containing microfilaria from a patient with no detectable microfilaria in the blood, are frequently observed. Two symptoms are both specific and frequent in infected subjects both with and without microfilaria in the blood: subconjunctival migration of an adult worm and elusive, migrating edemas of the hands, wrists and lower part of the forearm” [9]. “The index of subconjunctival filarial migration over the preceding year is particularly useful, because it correlates well with the microfilarial index but is more sensitive” (Figure 1). “Its determination involves precise questioning of the patient, which can be facilitated by the use of a demonstration chart, with diagrams and photographs” (see Figure 2).Open in a separate windowFigure 1Index of the subconjunctival migration of Loa loa adult worms and microfilarial index.Reproduced from Medicine Tropicale [3], released under CC BY 2.0 by Medicine Tropicale. IMSC = Indice de Migration Sous-Conjonctivale in French and Index of the SubConjunctival Migration in English. IM = Indice Microfilarien in French and Microfilarial Index in English.Open in a separate windowFigure 2Illustration of the passage of an adult worm (Loa loa) across the eye.This illustration (diagram and photograph) was made for presentation to patients questioned in endemic regions.The conclusion of this article was formulated as follows: “Screening for foci of filarial endemicity could be improved by the use of a simplified method and the validation of simple, inexpensive indices. Once these foci have been identified, a more precise evaluation can be carried out.”What is most astounding about the two WHO publications cited as the origin of this “new epidemiological index” [5], [6] is that the principal authors come from French-speaking African countries and/or work in or with this institution. They would therefore have been able to understand articles written in French. Furthermore, the WHO has a long-standing culture of multilingualism, particularly in English and French.Against this background, the rejection by the Bulletin of the World Health Organization and by other international journals published in English of an opinion article dealing with this issue and using the example of lymphatic filariasis does not seem to be justified, and is another illustration of “to be in English or not to be.”Indeed, filariasis due to Wuchereria bancrofti is systematically described as endemic in Congo and Gabon, two French-speaking countries, in non-specialist works on tropical medicine and in more specialist publications (WHO) despite a total absence of epidemiologic studies and/or confirmed case report over the last 30 years to prove it. What is certain is that no case was found when the last studies were conducted in these countries at the end of the 1970s and during the 1980s but unfortunately published in French. The studies that we carried out in the Congo as part of the National Project on Onchocerciasis and Other Filariases (between 1982 and 1987) confirmed the presence of four types of human filariasis: onchocerciasis, loaiasis, and the filariases caused by Mansonella perstans and M. streptocerca. There was a total absence of confirmed cases of lymphatic filariasis (bancroftosis). In this case, it is not a question of the attribution of merit for a particular “discovery”, but of basic knowledge of the geographic distribution of a scarcely studied disease, lymphatic filiariasis, in French Central Africa. Taking into account only publications in English, even older and poorly structured data, have been, in our opinion, a source of confusion and has led to false conclusions being drawn about the distribution range of this disease. This undoubtedly highlights the need to update knowledge by carrying out prospective studies (which seem to be underway), but these studies do not seem to be considered a matter of priority given the low levels of resources available and current health priorities.This has drawn us to publish this article in a French-language journal, but together with an entire translation into English [10]. Despite the bilingual nature of this publication, the international PubMed database identifies this article as being published in French, effectively ensuring that it will never be consulted, a classic “catch 22” situation! Indeed, this reference has never yet been cited by another author in a journal published in English. It may be that publication of an article in another language than English makes it more likely that it will not be cited, even if the authors of a subsequent article have access to the journal in which it was published and can understand the language used. Here, we begin to encroach on ethical problems and it is probably best not to delve too deeply. However, suffice it to say that the limited dissemination of publications in a language other than English may account for such equivocal attitudes.The problem is not a rivalry between French and English, but the confrontation between English and all other languages of the world. Moreover, the problem is undoubtedly worse for works published in non–Western European languages such as Chinese, Russian, and Japanese, which are arguably even less accessible.All things considered, this problem may seem a bit outdated, given that all “good authors” now publish exclusively in English. It certainly is outdated for most areas of medicine, where everything that is old is assigned to being nothing more than the history of medicine. But, surely, this should not be the case for NTDs, for which certain long-standing findings are every bit as important as what may be presented as new discoveries.One possibility would be for certain journals, such as PLOS NTDs, to include a specific heading permitting the publication of older studies that initially appeared in a language other than English (and are therefore currently little known). The texts included in this heading would essentially be English versions of these articles previously published in other languages, respecting the entirety of the original text.This would concern studies considered of importance because they highlight a point that remains unclear or describe an aspect considered innovative in a review but for which the originality of the article is due more to an incomplete reference list than to a true advance in knowledge. These articles should be judged in light of the knowledge and technical and methodological means available at the time at which they were initially published. Submission should be accompanied by a presentation of the problem, with details and explanatory comments, with submission at the initiative of the authors of the article in question or their students or sympathizers.  相似文献   

13.
Estimating the case-fatality risk (CFR)—the probability that a person dies from an infection given that they are a case—is a high priority in epidemiologic investigation of newly emerging infectious diseases and sometimes in new outbreaks of known infectious diseases. The data available to estimate the overall CFR are often gathered for other purposes (e.g., surveillance) in challenging circumstances. We describe two forms of bias that may affect the estimation of the overall CFR—preferential ascertainment of severe cases and bias from reporting delays—and review solutions that have been proposed and implemented in past epidemics. Also of interest is the estimation of the causal impact of specific interventions (e.g., hospitalization, or hospitalization at a particular hospital) on survival, which can be estimated as a relative CFR for two or more groups. When observational data are used for this purpose, three more sources of bias may arise: confounding, survivorship bias, and selection due to preferential inclusion in surveillance datasets of those who are hospitalized and/or die. We illustrate these biases and caution against causal interpretation of differential CFR among those receiving different interventions in observational datasets. Again, we discuss ways to reduce these biases, particularly by estimating outcomes in smaller but more systematically defined cohorts ascertained before the onset of symptoms, such as those identified by forward contact tracing. Finally, we discuss the circumstances in which these biases may affect non-causal interpretation of risk factors for death among cases.The case-fatality risk (CFR) is a key quantity in characterizing new infectious agents and new outbreaks of known agents. The CFR can be defined as the probability that a case dies from the infection. Several variations of the definition of “case” are used for different infections, as discussed in Box 1. Under all these definitions, the CFR characterizes the severity of an infection and is useful for planning and determining the intensity of a response to an outbreak [1,2]. Moreover, the CFR may be compared between cases who do and do not receive particular treatments as a way of trying to estimate the causal impact of these treatments on survival. Such causal inference might ideally be done in a randomized trial in which individuals are randomly assigned to treatments, but this is often not possible during an outbreak for logistical, ethical, and other reasons [3]. Therefore, observational estimates of CFR under different treatment conditions may be the only available means to assess the impact of various treatments.

Box 1. Definition of the CFR.

The CFR itself is an ambiguous term, as its definition and value depend on what qualifies an individual to be a “case.” Several different precise definitions of CFR have been used in practice, as have several imprecise ones. The infection-fatality risk (sometimes written IFR) defines a case as a person who has shown evidence of infection, either by clinical detection of the pathogen or by seroconversion or other immune response. Such individuals may or may not be symptomatic, though asymptomatic ones may go undetected. The symptomatic case-fatality risk (sCFR) defines a case as someone who is infected and shows certain symptoms. Infection in many outbreaks is given several gradations, including confirmed (definitive laboratory confirmation), probable (high degree of suspicion, by various clinical and epidemiologic criteria, without laboratory confirmation), and possible or suspected (lower degree of suspicion). This paper describes issues in estimating any of these risks or comparing them across groups, but does not go into the details of each possible definition.Furthermore, unlike risks commonly used in epidemiologic research (e.g., the 5-year mortality risk), the length of the period during which deaths are counted for the CFR is rarely explicit, probably because it is considered to be short enough to avoid ambiguity in the definition of CFR. However, a precise definition of the CFR would need to include the risk period, e.g., the 1-month CFR of Ebola. Clearly, the definition of CFR for a particular investigation should be specified as precisely as possible.However, observational studies conducted in the early phases of an outbreak, when public health authorities are appropriately concentrating on crisis response and not on rigorous study design, are challenging. A common problem is that disease severity of the cases recorded in a surveillance database will differ, perhaps substantially, from that of all cases in the population. This issue has arisen in the present epidemic of Ebola virus disease in West Africa and in many previous outbreaks and epidemics [49] and will continue to arise in future ones.Here we outline two biases that may occur when estimating the CFR in a population from a surveillance database, and three more biases that may occur when comparing the CFR between subgroups to estimate the causal effect of medical interventions. We also briefly consider the applicability of these biases to a different application: comparing the CFR across different groups of people, for example, by geography, sex, age, comorbidities, and other “unchangeable” risk factors. Such factors are “unchangeable” in the sense that they are not candidates for intervention in the setting of the outbreak, though some could, of course, change over longer timescales. The goal of estimating the CFR in groups defined by such unchangeable factors is not to understand the causal role of these factors in mortality, but to develop a predictive model for mortality that might be used to improve prognostic accuracy or identify disparities. Such predictions may be affected by survivorship bias and selection bias, but not by confounding, as we discuss.  相似文献   

14.

Background

The pathogenesis of rabies is associated with the inability to deliver immune effectors across the blood-brain barrier and to clear virulent rabies virus from CNS tissues. However, the mechanisms that facilitate immune effector entry into CNS tissues are induced by infection with attenuated rabies virus.

Methodology/Principal Findings

Infection of normal mice with attenuated rabies virus but not immunization with killed virus can promote the clearance of pathogenic rabies virus from the CNS. T cell activity in B cell–deficient mice can control the replication of attenuated virus in the CNS, but viral mRNA persists. Low levels of passively administered rabies virus–neutralizing antibody reach infected cells in the cerebellum of B cell–deficient mice but are not sufficient to mediate virus clearance. Production of rabies virus-specific antibody by B cells invading CNS tissues is required for this process, and a substantial proportion of the B cells that accumulate in the CNS of mice infected with attenuated rabies virus produce virus-specific antibodies.

Conclusions/Significance

The mechanisms required for immune effectors to enter rabies virus-infected tissues are induced by infection with attenuated rabies virus but not by infection with pathogenic rabies viruses or immunization with killed virus. T cell activities can inhibit rabies virus replication, but the production of rabies virus–specific antibodies by infiltrating B cells, as opposed to the leakage of circulating antibody across the BBB, is critical to elimination of the virus. These findings suggest that a pathogenic rabies virus infection may be treatable after the virus has reached the CNS tissues, providing that the appropriate immune effectors can be targeted to the infected tissues.  相似文献   

15.
The dengue fever epidemic in Guangzhou may have been affected by the Coronavirus Disease 2019 (COVID-19) pandemic. The number of dengue cases dropped drastically in 2020, and there have been only 2 local cases, suggesting that dengue has not become endemic in Guangzhou.

Guangzhou is located on the southeast coast of China and is the country’s third largest city. Since 1978, outbreaks of dengue fever have occurred intermittently in this city. In the past decade, the number of reported dengue cases reached more than 1,000 in 2013, 2014, 2018, and 2019, with 37,385 cases reported in 2014 alone. Therefore, dengue fever is a major public health concern in Guangzhou, and there is a continuing argument that it is endemic in Guangzhou [13].The numbers of dengue cases from 2017 to 2020 are shown in Table 1. In 2020, the total and local case numbers dropped dramatically compared to the previous years. With a high proportion of imported cases (n = 32, 94.12%), the proportion of local cases (n = 2, 5.88%) was considerably low in 2020. All the prevention and control strategies for dengue, including issuing public education messages, preventing further mosquito bites in patients, cleaning vector breeding sites, and using pesticides, were similar during these years. Additionally, dengue, as a mosquito-borne viral infectious disease, is closely related to mosquito density. The mosquito ovitrap index (MOI), which is the proportion of positive mosquito ovitraps, is usually used to indicate mosquito density. The MOI in 2017, 2018, and 2019 was 7.073 ± 1.016, 9.657 ± 1.307, and 8.464 ± 0.961, respectively. The average MOI was 8.398 ± 0.648 from 2017 to 2019 in Guangzhou. The MOI in 2020 was 7.135 ± 0.786, which remained at the median risk level. Therefore, the abnormal decline in dengue cases could not be attributed to the change in mosquito density in 2020.Table 1Numbers and percentages of dengue cases from 2017 to 2019.
Year2017201820192020
Total cases9441,2951,65534
Imported cases(percentage)69 (7.31%)96 (7.41%)270 (16.31%)32 (94.12%)
Local cases (percentage)875 (92.69%)1,199 (92.59%)1,385 (83.69%)2 (5.88%)
Open in a separate windowIn 2020, the 14-day quarantine in a designated hotel for international travelers to curb the spread of Coronavirus Disease 2019 (COVID-19) was an important public health intervention. People had to remain indoors except for medical care needs. All imported dengue cases were identified during their quarantine periods. No secondary case related to the imported cases was reported. This may be because Aedes albopictus, which is the major vector of dengue in Guangzhou, bites aggressively during the day outdoors. The chance of being bitten by A. albopictus was reduced by staying all day indoors. Moreover, some research revealed that viremia occurred 6 to 18 hours before symptoms appeared and lasted as long as 12 days [4]. After the 14-day quarantine, viremia had almost subsided. Therefore, imported dengue cases were unlikely to be transmitted. The impact of imported dengue cases was limited by the quarantine, which provided a rare opportunity to identify the local epidemic.The epidemiology investigation showed that the 2 local cases, who were living in the same building, had no travel history outside Guangzhou in 2020 and had symptoms successively. Two dengue virus serotype 2 (DENV-2) strains were isolated from them. The envelope gene sequences were obtained and deposited in GenBank under accession numbers MW295818 and MW345921. Reference sequences, which were downloaded from GenBank, and sequences of Guangzhou strains identified in the previous years, were used to construct a phylogenetic tree. The 2 isolated strains in 2020 were identical. The tree (Fig 1) shows that the 2 strains belonged to the Malaysia/Indian subcontinent genotype, which was the prevailing genotype in Guangzhou [5]. However, they were neither identical with nor derived from the Guangzhou strains obtained from the previous years. Using the Basic Local Alignment Search Tool in GenBank, the 2 strains were found to be highly similar to those identified in Zhejiang (China), Singapore, and Guangdong (China) in 2017. These results imply that the local cases may be secondary to some undiscovered cases imported from other cities in China, as no restriction and quarantine was imposed for domestic travels.Open in a separate windowFig 1Maximum-likelihood phylogenetic tree shows the evolutionary relationships of DENV-2 detected in the sera of 2 local cases along with 45 other sequences.The reference sequences are named using the GenBank accession number, country, and year. The sequences of strains isolated in Guangzhou are named using the GenBank accession number, year, and our lab number. Bootstrap support values are shown in the notes. Strains isolated in 2020 are indicated with a black triangle.When the impact of imported dengue cases was limited by quarantine, dengue did not spread in Guangzhou during 2020, with the MOI still at median risk level and without any changes in the prevention and control strategies. Moreover, serotype 1 had been prevalent in Guangzhou since 2011 [6,7]. However, there was no local infection of serotype 1 detected in 2020. These observations may provide further evidence that dengue fever is not endemic in Guangzhou.In conclusion, the number of dengue cases decreased during the COVID-19 epidemic in Guangzhou in 2020. Thus, we believe that dengue fever is not endemic in Guangzhou.  相似文献   

16.

Background

Rabies is a fatal viral encephalitic disease that is caused by lyssaviruses which can affect all mammals, including human and bats. In Europe, bat rabies cases are attributed to five different lyssavirus species, the majority of rabid bats being attributed to European bat 1 lyssavirus (EBLV-1), circulating mainly in serotine bats (Eptesicus serotinus). In France, rabies in bats is under surveillance since 1989, with 77 positive cases reported between 1989 and 2016.

Case presentation

In the frame of the bat rabies surveillance, an unusual mortality of serotine bats was reported in 2009 in a village in North-East France. Six juvenile bats from an E. serotinus maternity colony counting ~200 individuals were found to be infected with EBLV-1. The active surveillance of the colony by capture sessions of bats from July to September 2009 showed a high detection rate of neutralising EBLV-1 antibodies (≈ 50%) in the colony. Moreover, one out of 111 animals tested was found to shed viable virus in saliva, while lyssavirus RNA was detected by RT-PCR for five individuals.

Conclusion

This study demonstrated that the lyssavirus infection in the serotine maternity colony was followed by a high rate of bat rabies immunity after circulation of the virus in the colony. The ratio of seropositive bats is probably indicative of an efficient virus transmission coupled to a rapid circulation of EBLV-1 in the colony.
  相似文献   

17.

Introduction

In the continental US, four terrestrial mammalian species are reservoirs for seven antigenic rabies virus variants. Cross species transmission (CST) occurs when a rabies virus variant causes disease in non-reservoir species.

Methods

This study analyzed national surveillance data for rabies in terrestrial mammals. The CST rate was defined as: number of rabid non-reservoir animals/number of rabid reservoir animals. CST rates were analyzed for trend. Clusters of high CST rate counties were evaluated using space-time scanning statistics.

Results

The number of counties reporting a raccoon variant CST rate >1.0 increased from 75 in 1992 to 187 in 2011; counties with skunk variant CST rates >1.0 remained unchanged during the same period. As of 2011, for every rabid raccoon reported within the raccoon variant region, there were 0.73 cases of this variant reported in non-reservoir animals. Skunks were the most common non-reservoir animal reported with the raccoon rabies variant. Domestic animals were the most common non-reservoir animal diagnosed with a skunk rabies virus variant (n = 1,601). Cross species transmission rates increased fastest among domestic animals.

Conclusions

Cross species transmission of rabies virus variants into non-reservoir animals increases the risk of human exposures and threatens current advances toward rabies control. Cross species transmission in raccoon rabies enzootic regions increased dramatically during the study period. Pet owners should vaccinate their dogs and cats to ensure against CST, particularly in regions with active foci of rabies circulation. Clusters of high CST activity represent areas for further study to better understand interspecies disease transmission dynamics. Each CST event has the potential to result in a rabies virus adapted for sustained transmission in a new species; therefore further understanding of the dynamics of CST may help in early detection or prevention of the emergence of new terrestrial rabies virus variants.  相似文献   

18.

Purpose of Review

Onychomycosis is a growing global health problem. Proper diagnosis is important in order to avoid the risks of unnecessary treatments. This review summarizes the laboratory techniques currently available for the diagnosis of onychomycosis with the advantages and disadvantages of each test.

Recent Findings

Research is underway to prove the clinical utility of other diagnostic techniques such as PAS-stained nail biopsies and molecular tests as viable alternatives for the diagnosis of onychomycosis with promissory results.

Summary

Dermoscopy can be helpful to differentiate onychomycosis from traumatic onycholysis or true melanonychia; however, a definitive diagnosis requires the demonstration of the presence of the fungi. The combination of direct microscopy using potassium hydroxide and fungal culture is the gold standard for the diagnosis of onychomycosis; however, other alternatives, such as calcofluor white under fluorescence, PAS-stained nail biopsies, and molecular techniques have proven to be excellent methods with their advantages and limitations.
  相似文献   

19.

Objectives

Domestic dogs are the main reservoir of rabies virus (RABV) infection in Nigeria, thus surveillance of rabies in dog populations is crucial in order to understand the patterns of spread of infection and ultimately devise an appropriate rabies control strategy. This study determined the presence of lyssavirus antigen in brain tissues and anti-rabies antibodies in sera of apparently healthy and suspected-rabid dogs slaughtered for human consumption at local markets in South-Eastern Nigeria.

Results

Our findings demonstrated that 8.3% (n?=?23) of brain tissues were lyssavirus positive and 2.5% (n?=?25) of sera had rabies antibody levels as percentage blocking of 70% and above correlating with a cut-off value?≥?0.5 IU/mL in the fluorescent antibody neutralization test. There was an inverse correlation between lyssavirus positivity and rabies antibody levels confirming that infected individuals most often do not develop virus neutralizing antibodies to the disease. The low percentage of rabies antibodies in this dog population suggests a susceptible population at high risk to RABV infection. These findings highlight a huge challenge to national rabies programs and subsequent elimination of the disease from Nigeria, considering that majority of dogs are confined to rural communal areas, where parenteral dog vaccination is not routinely undertaken.
  相似文献   

20.

Background:

There is uncertainty about the optimal approach to screen for clinically important cervical spine (C-spine) injury following blunt trauma. We conducted a systematic review to investigate the diagnostic accuracy of the Canadian C-spine rule and the National Emergency X-Radiography Utilization Study (NEXUS) criteria, 2 rules that are available to assist emergency physicians to assess the need for cervical spine imaging.

Methods:

We identified studies by an electronic search of CINAHL, Embase and MEDLINE. We included articles that reported on a cohort of patients who experienced blunt trauma and for whom clinically important cervical spine injury detectable by diagnostic imaging was the differential diagnosis; evaluated the diagnostic accuracy of the Canadian C-spine rule or NEXUS or both; and used an adequate reference standard. We assessed the methodologic quality using the Quality Assessment of Diagnostic Accuracy Studies criteria. We used the extracted data to calculate sensitivity, specificity, likelihood ratios and post-test probabilities.

Results:

We included 15 studies of modest methodologic quality. For the Canadian C-spine rule, sensitivity ranged from 0.90 to 1.00 and specificity ranged from 0.01 to 0.77. For NEXUS, sensitivity ranged from 0.83 to 1.00 and specificity ranged from 0.02 to 0.46. One study directly compared the accuracy of these 2 rules using the same cohort and found that the Canadian C-spine rule had better accuracy. For both rules, a negative test was more informative for reducing the probability of a clinically important cervical spine injury.

Interpretation:

Based on studies with modest methodologic quality and only one direct comparison, we found that the Canadian C-spine rule appears to have better diagnostic accuracy than the NEXUS criteria. Future studies need to follow rigorous methodologic procedures to ensure that the findings are as free of bias as possible.A clinically important cervical spine injury is defined as any fracture, dislocation or ligamentous instability detectable by diagnostic imaging and requiring surgical or specialist follow-up.1,2 These injuries can have disastrous consequences including spinal cord injury and death if the diagnosis is delayed or missed.3 Despite the low prevalence (< 3%) of clinically important cervical spinal injury following blunt trauma (e.g., motor vehicle collision), accurate diagnosis is imperative for safe, effective management.4 Currently, uncertainty exists about the optimal diagnostic approach. Some guidelines5,6 advocate using screening tools to identify patients with a higher likelihood of clinically important cervical spinal injury; these patients are then sent for imaging to establish the diagnosis. In other more conservative settings, all patients with blunt trauma are sent for imaging. The first approach, involving screening, is arguably preferable because it optimizes resources and time, while reducing unnecessary costs, radiation exposure and psychological stress for the patient.7 For screening to be safe and effective, the screening tools must have high sensitivity, a low negative likelihood ratio and a low rate of false positives. This assures clinicians that a clinically important cervical spine injury is unlikely and reduces the number of referrals for imaging.Clinical decision rules synthesize 3 or more findings from the patient’s history, physical examination or simple diagnostic tests to guide diagnostic and treatment decisions.8,9 Two clinical decision rules, the Canadian C-spine rule2 and the National Emergency X-Radiography Utilization Study (NEXUS; Box 1),10 are available to assess the need for imaging in patients with cervical spine injury following blunt trauma. These rules aim to reduce unnecessary imaging by reserving these investigations for patients with a higher likelihood of clinically important cervical spinal injury. Developed independently and validated using large cohorts of patients, these 2 decision rules are recommended in many international guidelines.5,11,12 However, no consensus exists as to which rule should be endorsed.1214 Therefore, the purpose of our systematic review was to describe the quality of research evaluating the Canadian C-spine rule and NEXUS; describe the diagnostic accuracy of the Canadian C-spine rule and NEXUS; and compare the diagnostic accuracy of the Canadian C-spine rule to that of NEXUS.

Box 1:

National Emergency X-Radiography Utilization Study (NEXUS) low-risk criteria10

Cervical spine radiography is indicated for patients with neck trauma unless they meet ALL of the following criteria:
  • No posterior midline cervical-spine tenderness
  • No evidence of intoxication
  • A normal level of alertness (score of 15 on the Glasgow Coma Scale)
  • No focal neurologic deficit
  • No painful distracting injuries
  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号