12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Lymphatic pathology in asymptomatic and symptomatic children with Wuchereria bancrofti infection in children from Odisha, India and its reversal with DEC and albendazole treatment

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Once interruption of transmission of lymphatic filariasis is achieved, morbidity prevention and management becomes more important. A study in Brugia malayi filariasis from India has shown sub-clinical lymphatic pathology with potential reversibility. We studied a Wuchereria bancrofti infected population, the major contributor to LF globally.

          Methods

          Children aged 5–18 years from Odisha, India were screened for W. bancrofti infection and disease. 102 infected children, 50 with filarial disease and 52 without symptoms were investigated by lymphoscintigraphy and then randomized to receive a supervised single oral dose of DEC and albendazole which was repeated either annually or semi-annually. The lymphatic pathology was evaluated six monthly for two years.

          Findings

          Baseline lymphoscintigraphy showed abnormality in lower limb lymphatics in 80% of symptomatic (40/50) and 63·5% (33/52) of asymptomatic children. Progressive improvement in baseline pathology was seen in 70·8, 87·3, 98·6, and 98·6% of cases at 6, 12, 18, and 24 months follow up, while in 4·2, 22·5, 47·9 and 64·8%, pathology reverted to normal. This was independent of age (p = 0·27), symptomatic status (p = 0·57) and semi-annual/bi-annual dosing (p = 0·46). Six of eleven cases showed clinical reduction in lymphedema of legs.

          Interpretation

          A significant proportion of a young W. bancrofti infected population exhibited lymphatic pathology which was reversible with annual dosage of DEC and albendazole. This provides evidence for morbidity prevention & treatment of early lymphedema. It can also be used as a tool to improve community compliance during mass drug administration.

          Trial registration

          ClinicalTrials.gov No CTRI/2013/10/004121

          Author summary

          Infection with lymphatic filarial parasites usually occurs early in childhood in endemic areas, but clinical signs appear much later. Reversal of lymphatic pathology has been shown with the much less common Brugia malayi infection using DEC and albendazole and there is scarce evidence whether the same occurs with bancroftian filariasis using the above drugs. We designed this study to look for prevalence of lymphatic pathology in children with and without clinical signs of infection and to observe the effect on the pre-treatment pathology using DEC and albendazole given once or twice a year. We have shown, using lymphoscintigraphy, that lymphatic vessel changes occur very early in infection, and treatment can reverse these changes, even when clinical symptoms are already apparent. This has important implications for lymphedema prevention, case management and for advocacy in the Lymphatic Filariasis Elimination Program. It also strengthens the previous evidence of benefit of treatment on early lymphedema management.

          Related collections

          Most cited references31

          • Record: found
          • Abstract: found
          • Article: not found

          Strategies and tools for the control/elimination of lymphatic filariasis.

          Lymphatic filariasis infects 120 million people in 73 countries worldwide and continues to be a worsening problem, especially in Africa and the Indian subcontinent. Elephantiasis, lymphoedema, and genital pathology afflict 44 million men, women and children; another 76 million have parasites in their blood and hidden internal damage to their lymphatic and renal systems. In the past, tools and strategies for the control of the condition were inadequate, but over the last 10 years dramatic research advances have led to new understanding about the severity and impact of the disease, new diagnostic and monitoring tools, and, most importantly, new treatment tools and control strategies. The new strategy aims both at transmission control through community-wide (mass) treatment programmes and at disease control through individual patient management. Annual single-dose co-administration of two drugs (ivermectin + diethylcarbamazine (DEC) or albendazole) reduces blood microfilariae by 99% for a full year; even a single dose of one drug (ivermectin or DEC) administered annually can result in 90% reductions; field studies confirm that such reduction of microfilarial loads and prevalence can interrupt transmission. New approaches to disease control, based on preventing bacterial superinfection, can now halt or even reverse the lymphoedema and elephantiasis sequelae of filarial infection. Recognizing these remarkable technical advances, the successes of recent control programmes, and the biological factors favouring elimination of this infection, the Fiftieth World Health Assembly recently called on WHO and its Member States to establish as a priority the global elimination of lymphatic filariasis as a public health problem.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The global programme to eliminate lymphatic filariasis.

            E Ottesen (2000)
            Ten years ago, no one foresaw that in the year 2000 there would be a Global Programme to Eliminate Lymphatic Filariasis (GPELF) that is already 2 years old, active in 18 of the 80 endemic countries, and operating under a wholly new paradigm in public health - a paradigm affirming that public/private sector partnerships are essential in sharing both responsibilities and responses to global health problems. What has driven the LF Elimination Programme to this point? Where it is now headed? What will be required to sustain its momentum? What will its impact be? These are the issues addressed below.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Doxycycline Improves Filarial Lymphedema Independent of Active Filarial Infection: A Randomized Controlled Trial

              Worldwide, 40 million persons are disabled owing to filariasis-related morbidity, with 15 million suffering from lymphedema (LE) or elephantiasis and 25 million with hydrocele. Filarial LE is caused by infection with the lymphatic filarial nematodes, Wuchereria bancrofti, Brugia malayi, and Brugia timori and occurs most commonly in the legs. Patients experience a gradual and progressive development in the severity of clinical LE, graded 1–7 (Supplementary Figure 1), leading to severe disability, loss of productivity, and social stigmatization [1]. Patients with LE also experience “acute attacks” or acute dermatolymphangioadenitis, arising through secondary microbial infection acquired through lesions in the skin [2]. The goals of the Global Programme to Eliminate Lymphatic Filariasis (GPELF) are (1) to interrupt transmission by reduction of microfilaremia levels using mass drug administration (MDA) of filaricidal drugs and (2) to provide morbidity management for those who suffer from clinical manifestations associated with lymphatic filariasis (LF) [3–5]. The first goal has been successfully approached in areas having used diethylcarbamazine and albendazole for mass drug treatment for ≥5 years but has been met less successfully in Africa, where ivermectin and albendazole have been administered [6]. During the first 10 years of the GPELF, prevention of new morbidity has been impressive, with an estimated 22 million persons protected from LF infection and disease, accounting for economic savings of US $24.2 billion [4]. However, the goal to reduce existing morbidity associated with chronic filarial disease has not scaled up as rapidly as MDA, with only 33% of endemic countries introducing morbidity management in the first decade of the GPELF [1]. Current morbidity management strategies rely on improving hygiene and skin care of affected limbs, with limb elevation, exercise, and the use of topical antibiotics and antifungal creams, which reduces the frequency of acute attacks and can help arrest the development of LE [3]. We have shown elsewhere that 6 weeks of doxycycline results in a significant amelioration of LE severity in patients with active infection of W. bancrofti [7]. The present study was designed to address whether these benefits of a 6-week course of doxycycline (200 mg/d) could be extended to patients negative for circulating filarial antigen (CFA). In addition, we tested to determine whether a similar course of amoxicillin was also effective against LE. Our results demonstrate that doxycycline improves mild to moderate LE over 2 years. Importantly, CFA-negative patients also demonstrated significant improvement in LE, showing that the activity of doxycycline is not confined to patients with active infection. These results strongly promote the use of doxycycline as a new strategy for improved morbidity management of LF. MATERIALS AND METHODS The trial was approved by the Committee on Human Research, Publication and Ethics at the Kwame Nkrumah University of Science and Technology, Kumasi, Ghana. The study conformed to the principles of the Declaration of Helsinki 1964 (amended most recently in 2004). The trial registration number is ISRCTN 90861344. Participants and Study Area This trial was conducted in 21 endemic communities in the Nzema East and Ahanta West districts in the western region of Ghana [7–9], where MDA was started in 2001. Written informed consent was obtained from all participants. Eligible individuals were adults of both sexes aged 18–60 years with LE stage 1–5 according to the staging scheme by Dreyer [5], with a minimum body weight of 40 kg and no requirement for chronic medication. Exclusion criteria included LE stage 6 or 7; abnormal gamma-glutamyltransferase, alanine aminotransferase, or creatinine levels; pregnancy; breast-feeding; intolerance to doxycycline or amoxicillin; and alcohol or drug abuse. A clinician (S. M.) experienced in the symptoms of LE examined all consenting patients and performed the staging as well as ultrasonography at pretreatment and follow-up examinations. Randomization and Masking This was a randomized trial; the design was placebo controlled for the comparison of doxycycline and placebo, with an additional open amoxicillin arm. Randomization was carried out using computer-generated software. Blinding (masking) was ensured by exclusion of study personnel performing clinical or laboratory analyses from randomization or tablet distribution. To avoid an influence on results caused by the open amoxicillin arm, the personnel administering the treatment (A. B. D. and Y. M. D.) did not take part in follow-up examinations. A total of 162 patients were randomly allocated into 3 treatment arms: (1) amoxicillin, (2) doxycycline, and (3) placebo treatment, each with 54 patients. Before randomization, participants were stratified into those with and those without active infection (CFA positive and CFA negative). These 2 subgroups were separately randomized, resulting in 6 final treatment groups (Figure 1, Supplementary Figure 2A and B ). Figure 1. Participant flow. Flow chart of all assessed volunteers (circulating filarial antigen positive and negative). Abbreviations: CFA, circulating filarial antigen; USG, ultrasonography. Treatment and Follow-up Participants received doxycycline (2 100-mg capsules daily), matching placebo, or amoxicillin (2 500-mg tablets daily) for a total of 6 weeks. Treatment was directly observed daily. Adverse events were recorded in the case report forms, and patients were seen by the trial clinician (A. B. D.). Two rounds of single-dose ivermectin and albendazole were distributed during the clinical trial within the scope of the annual MDA. Follow-up time points were 3, 12, and 24 months after treatment onset. Foot Care Training and Monitoring of Hygiene Measures All patients received soap, towels, and plastic bowls for washing their legs and were trained in foot hygiene, according to the booklet “New Hope” for persons with LE [5]. LE Staging Staging of LE was performed according to the “Basic Lymphedema Management” guidelines established by G. Dreyer and colleagues [5] (Supplementary Figure 1). To allow comparisons of LE staging in patients with either one or both legs affected, the following approach was employed: (1) if only one leg showed LE, this leg was analyzed; (2) if both legs were affected, the leg with the lower stage is reported; (3) if both legs were affected showing equal stages, either the left or the right leg was analyzed after computer-generated randomization. Monitoring of Acute Attacks Acute attacks were defined as pain, lymph node swelling (femoral and/or inguinal), fever, and peeling of the skin on the affected leg after the resolving of the febrile attack (Supplementary Figure 3). The history regarding acute attacks was evaluated based on booklets kept by the patients and questionnaires filled out by the research team at every visit. Each study participant was asked in detail about the number and duration of attacks, fever, lymph node swelling, and peeling of the skin to differentiate general pain in affected legs from genuine acute attacks. Microfilaremia, Antigenemia and Vascular Endothelial Growth Factors For quantification of microfilariae in the blood the Whatman Nucleopore filter method was applied using 10-mL samples of night blood before treatment and at follow-up. Levels of W. bancrofti CFA were determined using the TropBio enzyme-linked immunosorbent assay (ELISA) test (TropBio) [7]. Levels of vascular endothelial growth factor (VEGF) C and soluble VEGF receptor (VEGFR) 3 were measured from plasma samples using Quantikine or DuoSet ELISA kits, respectively (R&D systems). Circumference Measurement of Legs Leg circumferences were determined using a tape measure. Measurements were performed at 10 cm from the large toe and 12, 20, and 30 cm from the sole of the foot, as described elsewhere [10, 11]. Averages of the 4 measurements were determined before treatment and at follow-up. Ultrasound Examinations of the Ankles Ultrasonography was performed between 2 and 7 PM using a SonoSite 180 PLUS hand-carried ultrasound system (SonoSite) equipped with a 38-mm 5–10-MHz linear-array transducer [12]. Patients were scanned sitting with stretched legs and feet perpendicular to the legs. The transducer was positioned at the top of the malleolus (ankle) and kept at a 90° angle to the skin surface in transverse sections. The head of tibia or fibula had to be visible, and the malleolus had to appear as a sharp line (Supplementary Figure 1) to permit reproducibility. The thickness of the tissue (subcutis, dermis, and epidermis) was measured from the malleolus to the skin surface. Lateral and medial malleoli of both legs were measured before treatment and at follow-up. Statistical Analysis To compare between the treatment arms, the intraindividual differences in staging or measurements between the respective follow-up and pretreatment findings were first calculated. Kruskal-Wallis tests followed by Mann-Whitney U tests or Fisher's exact test were used to compare between the treatment arms. The Wilcoxon signed rank test or the McNemar test were chosen for comparing pretreatment with follow-up results in one treatment arm. Correlations were done using the Spearman rank test. Differences between the treatment arms in the survival curve (Kaplan-Meier) were analyzed with the log-rank test. To confirm the results of the per-protocol analyses (patients who completed the treatment and were present for the respective follow-up visit) an intention-to-treat (ITT) analysis was carried out with all patients who started treatment (missing values were filled in using the "last observation carried forward" method). Analyses were done using PASW statistics 18.0 software (IBM) and SAS 9.2 software (SAS). RESULTS Participants From a recruitment pool of 205 patients with LE, 162 patients were stratified according to CFA status (Figure 1, Supplementary Figure 2A and B ). At the start of treatment, 21 patients abandoned participation, although they had explicitly agreed to participate and had been randomized, and 13 participants stated that they had not given correct information about age or breast-feeding on recruitment. Along with these 34 patients, another 9 dropped out during treatment for reasons unrelated to the study drugs. Baseline Data Of the 119 patients who completed the treatment, 46 (39%) were CFA positive and 73 (61%) were CFA negative. Most of the patients had LE stage 2 (44.5%) or 3 (47.9%). The mean age was 47.7 ± 10.8 years. CFA-negative patients had on average a longer history of LE development than CFA-positive patients (P = .029). There was no difference among the treatment arms regarding compliance with MDA (Table 1). Table 1. Baseline Data Variable Total Doxycycline Amoxicillin Placebo P Value Patients (male/female), No. 119 (34/85) 46 (10/36) 35 (9/26) 38 (15/23) .198a  CFA positive 46 (17/29) 19 (5/14) 13 (6/7) 14 (6/8) .471a  CFA negative 73 (17/56) 27 (5/22) 22 (3/19) 24 (9/15) .153a  LE stage 1 2 (0/2) 1 (0/1) 1 (0/1)  0  LE stage 2 53 (16/37) 20 (5/15) 18 (4/14) 15 (7/8)  LE stage 3 57 (15/42) 23 (4/19) 15 (5/10) 19 (6/13)  LE stage 4 1 (0/1) 1 (0/1)  0  0  LE stage 5 6 (3/3) 1 (1/0) 1 (0/1) 4 (2/2) Age, mean ± SD, years 47.7 ± 10.8 46.1 ± 11.6 51.3 ± 9.1 46.3 ± 10.9 .045b  CFA positive 49.7 ± 12.1 45.5 ± 13.9 56.0 ± 5.6 49.4 ± 12.1 .045b  CFA negative 46.5 ± 9.8 46.5 ± 9.9 48.6 ± 9.7 44.5 ± 9.9 .304b Duration of LE, mean ± SD, years 13.8 ± 12.3 13.7 ± 11.8 14.6 ± 14.9 13.3 ± 10.5 .893b  CFA positive 11.5 ± 12.7 9.3 ± 10.2 12.2 ± 16.5 13.7 ± 12.3 .475b  CFA negative 15.3 ± 11.9 16.9 ± 11.9 16.0 ± 14.1 13.0 ± 9.5 .554b MDA/total, No. (%)  2006 87/117 (74) 31/46 (67.4) 28/34 (82.4) 28/37 (75.7) .329a  2007 85/115 (74) 30/44 (68.2) 27/34 (79.4) 28/37 (75.7) .535a  2008 75/109 (69) 25/41 (61.0) 24/31 (77.4) 26/37 (70.3) .347a Abbreviations: CFA, circulating filarial antigen; LE, lymphedema; MDA, mass drug administration (ivermectin plus albendazole); SD, standard deviation. a Fisher's exact test. b Kruskal-Wallis test. Primary Outcome Analysis LE Staging Figures 2 A–C show changes in LE stages before treatment compared with 12 and 24 months after treatment. The affected legs of patients in the doxycycline group reverted to a lower stage at 12 and 24 months (P = .002; Table 2), whereas they progressed to a higher stage in the amoxicillin and placebo groups (P = .012 and P = .001 respectively). Comparing all groups at 24 months (Figure 2 A), there was a difference between doxycycline and amoxicillin groups (P  0 denote an increase to higher LE stages. Abbreviation: LE, lymphedema. Figure 3. Kaplan-Meier curves showing occurrence of acute attacks after treatment end for each treatment arm. Arrows denote follow-up time points. The following significant differences between the 3 treatment arms were detected at 3 months: amoxicillin versus doxycycline (P = .018) and amoxicillin versus placebo (P = .007); at 12 months: doxycycline versus placebo (P = .012) and amoxicillin versus placebo (P = .007); and at 24 months: doxycycline versus placebo (P = .007). Figure 4. Reduction in skin thickness at the ankles, as analyzed by ultrasound. Box plots show differences in skin thickness at the ankles at 24 months compared to pretreatment (P = .001 for overall difference between the 3 treatment arms; Kruskal-Wallis test). Doxycycline was able to improve conditions in 36.6% of patients and halt progression in 58.5%; in 4.9% the legs became worse (Table 3). This improvement was significantly superior to that seen with amoxicillin or placebo (P  9 years old when LE begins) and pregnant women may be manageable in patients with LE. Tolerability and adverse events were the same in all treatment arms, which, together with our experience in all previous trials [7, 19–22], shows that doxycycline is well tolerated and safe. In conclusion, this trial clearly demonstrates that doxycycline is beneficial in reverting or halting the progression of early stages of filarial LE, regardless of whether there is still active infection. These findings lead us to recommend that individuals with filarial LE stage 1–3 should take a course of doxycycline (200 mg/d) for 6 weeks every other year, or maybe even yearly, and that doxycycline should be considered as a new tool to improve morbidity management of LE. Supplementary Data Supplementary materials are available at Clinical Infectious Diseases online (http://cid.oxfordjournals.org). Supplementary materials consist of data provided by the author that are published to benefit the reader. The posted materials are not copyedited. The contents of all supplementary data are the sole responsibility of the authors. Questions or messages regarding errors should be addressed to the author. Supplementary Data
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Negl Trop Dis
                PLoS Negl Trop Dis
                plos
                plosntds
                PLoS Neglected Tropical Diseases
                Public Library of Science (San Francisco, CA USA )
                1935-2727
                1935-2735
                23 October 2017
                October 2017
                : 11
                : 10
                : e0005631
                Affiliations
                [1 ] Director of Medical Research, IMS & Sum Hospital, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
                [2 ] Regional Medical Research Centre, Chandrasekhapur, Bhubaneswar, Odisha, India
                [3 ] Utkal Institute of Medical Sciences, Bhubaneswar, Odisha, India
                [4 ] Apollo Hospital, Bhubaneswar, Odisha, India
                [5 ] Universiti Sains Malaysia, Penang, Malaysia
                [6 ] Tropical Projects, Hitchin, United Kingdom
                University Hospital Bonn, GERMANY
                Author notes

                The authors have declared that no competing interests exist.

                • Conceptualization: SKK BD JH.

                • Data curation: BD.

                • Formal analysis: BD.

                • Funding acquisition: SKK.

                • Investigation: SKK BD BKD BKA.

                • Methodology: SKK BD JH BKD BKA CPR.

                • Project administration: SKK.

                • Resources: BD SKK.

                • Supervision: SKK CPR JH.

                • Validation: BD SKK.

                • Visualization: SKK BD JH.

                • Writing – original draft: SKK BD.

                • Writing – review & editing: BD SKK JH.

                ‡ SKK and BD share first authorship on this work.

                Author information
                http://orcid.org/0000-0001-7047-5106
                Article
                PNTD-D-16-01951
                10.1371/journal.pntd.0005631
                5667936
                29059186
                bd22a7e4-3291-4f74-b5e1-7e8c572c3139
                © 2017 Kar et al

                This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 25 October 2016
                : 10 May 2017
                Page count
                Figures: 10, Tables: 6, Pages: 19
                Funding
                Funded by: funder-id http://dx.doi.org/10.13039/100000865, Bill and Melinda Gates Foundation;
                Award ID: 43922
                Award Recipient :
                This work was funded by Bill & Melinda Gates Foundation Grant number 43922 to Task Force for Global Health Lymphatic Filariasis Support Centre. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Biology and Life Sciences
                Organisms
                Eukaryota
                Animals
                Invertebrates
                Nematoda
                Wuchereria
                Wuchereria Bancrofti
                Medicine and Health Sciences
                Diagnostic Medicine
                Diagnostic Radiology
                Lymphoscintigraphy
                Research and Analysis Methods
                Imaging Techniques
                Diagnostic Radiology
                Lymphoscintigraphy
                Medicine and Health Sciences
                Radiology and Imaging
                Diagnostic Radiology
                Lymphoscintigraphy
                Medicine and Health Sciences
                Diagnostic Medicine
                Signs and Symptoms
                Edema
                Lymphedema
                Medicine and Health Sciences
                Pathology and Laboratory Medicine
                Signs and Symptoms
                Edema
                Lymphedema
                Biology and Life Sciences
                Anatomy
                Musculoskeletal System
                Limbs (Anatomy)
                Legs
                Medicine and Health Sciences
                Anatomy
                Musculoskeletal System
                Limbs (Anatomy)
                Legs
                People and Places
                Geographical Locations
                Asia
                India
                Medicine and Health Sciences
                Health Care
                Health Statistics
                Morbidity
                Medicine and Health Sciences
                Pediatrics
                Pediatric Infections
                Biology and Life Sciences
                Organisms
                Eukaryota
                Animals
                Invertebrates
                Nematoda
                Brugia
                Brugia Malayi
                Custom metadata
                vor-update-to-uncorrected-proof
                2017-11-02
                All relevant data are within the paper and its Supporting Information files.

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article