1
|
Rijaul SKK, Maity N, Konar A, Hazra S. Topical Dexamethasone Counters Intravitreal Ivermectin-Induced Ocular Toxicity in a Rabbit Model. Curr Eye Res 2024; 49:750-758. [PMID: 38501588 DOI: 10.1080/02713683.2024.2330520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
PURPOSE Systemic use of Ivermectin has been reported to incite blindness in humans and veterinary patients. This study was designed to investigate the systemic and intravitreal effect of Ivermectin on ocular and retinal health and its attenuation with topical Dexamethasone. METHODS Systemic injection of Ivermectin@ 1.6 mg/kg S/C was administered, thrice a week for three weeks to New Zealand White rabbits (N = 4) with and without topical drops of Verapamil (N = 4). Pre and post-treatment ocular examination was conducted. At the end of three weeks the eyes were collected for histopathology.0.2 ml of Ivermectin solution (1.6 mg/ml) was injected intravitreally in one eye of the rabbit (N = 8), Half the rabbits received 0.1% dexamethasone drops thrice daily for 7 days, while the controls received PBS. Pre and post-treatment, detailed examination was conducted, which included the Schirmer Tear test, Fluorescein staining, Intraocular pressure, slit lamp biomicroscopy and fundus photography. The retina was harvested for histopathological and tunnel assay. RESULTS Systemic therapy with Ivermectin, with and without Verapamil did not incite any adverse response in the eye. Intravitreal Ivermectin evoked severe uveitis 4/4, cataract 3/4, corneal erosion 3/4 eyes and severe inflammatory response. Eyes that received dexamethasone were rescued from the adverse changes as demonstrated clinically, by histopathology and prevention of apoptosis. CONCLUSIONS Intravitreal Ivermectin incites severe inflammatory response. Topical dexamethasone counters the ocular toxicity incited by Ivermectin.
Collapse
Affiliation(s)
- S K K Rijaul
- Department of Veterinary Surgery & Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | - Namrata Maity
- Department of Veterinary Surgery & Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | | | - Sarbani Hazra
- Department of Veterinary Surgery & Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| |
Collapse
|
2
|
Hanson WH, Samuels CJ, Woods CL, Henderson KS. Evaluating a Reduction in Treatment Duration of Ivermectin Diet for Fur Mite ( Radfordia affinis) Eradication in Mice. Comp Med 2024; 74:167-172. [PMID: 39107940 PMCID: PMC11267440 DOI: 10.30802/aalas-cm-24049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/08/2024] [Accepted: 03/28/2024] [Indexed: 08/10/2024]
Abstract
Murine fur mites are commonly excluded in modern research animal programs, yet infestations continue to persist due to challenges in detection and control. Because all diagnostic methods and treatment options have limitations, programs must make many operational decisions when trying to eradicate these ectoparasites. The primary aim of this study was to assess various durations of treatment time with an ivermectin-compounded diet in eliminating Radfordia affinis in mice as determined by PCR testing and pelt examination. A shorter treatment duration would be highly advantageous as compared with the current regimen of 8 wk as it would minimize cost and time for animal management programs, impediments to research, and ivermectin drug effects on infested animals. Five experimental groups of R. affinis -positive mice received dietary ivermectin for 0, 2, 4, 6, or 8 wk. A fur mite-negative, naïve mouse was added to each group every 8 wk to perpetuate the infestation and amplify any remaining populations of fur mites. At 16 wk after the respective treatment end, PCR testing was performed for all treated groups in conjunction with the positive control group (no treatment). Visual examination of pelts for mites and eggs via direct microscopy was also performed at each time point. All treated mice were free of R. affinis at 16 wk after the end of treatment as confirmed by both PCR testing and pelt examination. These findings indicate that a dietary ivermectin treatment duration of as little as 2 wk is effective in eliminating R. affinis, making successful eradication initiatives more achievable.
Collapse
Affiliation(s)
- Wai H Hanson
- Division of Animal Resources, Emory University, Atlanta, Georgia; and
| | - Cayden J Samuels
- Division of Animal Resources, Emory University, Atlanta, Georgia; and
| | - Cheryl L Woods
- Research Animal Diagnostic Services, Charles River Laboratories, Wilmington, Massachusetts
| | - Kenneth S Henderson
- Research Animal Diagnostic Services, Charles River Laboratories, Wilmington, Massachusetts
| |
Collapse
|
3
|
Gordon CA, Utzinger J, Muhi S, Becker SL, Keiser J, Khieu V, Gray DJ. Strongyloidiasis. Nat Rev Dis Primers 2024; 10:6. [PMID: 38272922 DOI: 10.1038/s41572-023-00490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/27/2024]
Abstract
Strongyloidiasis is a neglected tropical disease caused primarily by the roundworm Strongyloides stercoralis. Strongyloidiasis is most prevalent in Southeast Asia and the Western Pacific. Although cases have been documented worldwide, global prevalence is largely unknown due to limited surveillance. Infection of the definitive human host occurs via direct skin penetration of the infective filariform larvae. Parasitic females reside in the small intestine and reproduce via parthenogenesis, where eggs hatch inside the host before rhabditiform larvae are excreted in faeces to begin the single generation free-living life cycle. Rhabditiform larvae can also develop directly into infectious filariform larvae in the gut and cause autoinfection. Although many are asymptomatic, infected individuals may report a range of non-specific gastrointestinal, respiratory or skin symptoms. Autoinfection may cause hyperinfection and disseminated strongyloidiasis in immunocompromised individuals, which is often fatal. Diagnosis requires direct examination of larvae in clinical specimens, positive serology or nucleic acid detection. However, there is a lack of standardization of techniques for all diagnostic types. Ivermectin is the treatment of choice. Control and elimination of strongyloidiasis will require a multifaceted, integrated approach, including highly sensitive and standardized diagnostics, active surveillance, health information, education and communication strategies, improved water, sanitation and hygiene, access to efficacious treatment, vaccine development and better integration and acknowledgement in current helminth control programmes.
Collapse
Affiliation(s)
- Catherine A Gordon
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia.
- Faculty of Medicine, University of Queensland, St Lucia, Brisbane, Queensland, Australia.
| | - Jürg Utzinger
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Stephen Muhi
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- The University of Melbourne, Department of Microbiology and Immunology, Parkville, Victoria, Australia
| | - Sören L Becker
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
- Institute of Medical Microbiology and Hygiene, Saarland University, Homburg/Saar, Germany
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Virak Khieu
- National Centre for Parasitology, Entomology and Malaria Control, Ministry of Health, Phnom Penh, Cambodia
| | - Darren J Gray
- Population Health Program, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| |
Collapse
|
4
|
Abstract
Ivermectin is an antiparasitic drug that has been used as an alternative for prophylaxis and treatment of COVID-19 infection. The adverse effects from supratherapeutic doses of ivermectin can include non-neurological and neurological symptoms. In this study, we report the case of a 52-year-old Filipino male with newly diagnosed diabetes mellitus who developed a subacute history of fever, cough, and generalized weakness, causing him to self-medicate with supratherapeutic doses of ivermectin and thereafter subsequently developed a decrease in sensorium, restlessness, and complex visual hallucinations. Significant laboratory examinations showed hyperglycemia, mild hyponatremia, positive SARS-CoV2 reverse transcriptase polymerase chain reaction test, and bilateral pneumonia on chest radiograph. He was subsequently started on antibiotics, a high-flow nasal cannula, and given two doses of activated charcoal. During the first 24 hours of hospital admission, there was a significant improvement in the patient's sensorium with a resolution of restlessness and visual hallucinations. During the rest of the hospitalization, his respiratory symptoms improved, and he was subsequently discharged. Clinical outcome in our patient after administration of activated charcoal and completion of antibiotics showed an overall improvement in symptoms and without any neurologic sequelae.
Collapse
Affiliation(s)
- Jon Stewart H Dy
- Institute for Neurosciences, St. Luke's Medical Center, Quezon City, PHL
| | - Dan N Juangco
- Institute for Neurosciences, St. Luke's Medical Center, Quezon City, PHL
| |
Collapse
|
5
|
Asiag N, Chai O, Yodovner S, Ruggeri M, Rapaport K, Baneth G, Nachum-Biala Y, Konstantin L, Rojas A, Brenner O, Bruchim Y, Shamir MH. Evaluation of a treatment protocol in dogs with intraspinal spirocercosis. J Am Vet Med Assoc 2022; 261:384-390. [PMID: 36476412 DOI: 10.2460/javma.22.09.0401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To evaluate the efficiency and safety of a doramectin-based treatment protocol in dogs affected by intraspinal spirocercosis (Spirocerca lupi). ANIMALS Client-owned dogs that were admitted to a veterinary hospital during 2021 to 2022 with acute onset of neurological signs and diagnosed with intraspinal spirocercosis. All dogs underwent complete neurological evaluation, CSF analysis, PCR confirmation of CNS S lupi infection, and follow-up evaluation of at least 6 months. PROCEDURES Upon diagnosis, dogs were treated with doramectin at a dose of 400 μg/kg, SC, q 24 h for 3 consecutive days, followed by the same dose once a week for 6 weeks. Prednisone was administered at a dose of 1 mg/kg, PO, q 24 h and tapered every 3 days. Antimicrobial clindamycin was administered at a dose of 12.5 mg/kg, PO, q 12 h for 7 days to reduce the risk of secondary spinal cord infection. Short- and long-term outcomes (1 week to 56 months) were recorded. RESULTS 8 dogs fulfilled the inclusion criteria, 7 of which presented with neurological deficits and 1 with cervical pain. Initiation of treatment was associated with stopping the deterioration in 7 of 8 dogs. Seven dogs improved and 6 recovered ambulation. One dog was euthanized due to lack of improvement. Six of the recovered dogs were still ataxic on the last follow-up examination at 6 to 56 months. No adverse effects of the drug were noted. CLINICAL RELEVANCE Frequent administration of doramectin was found to be safe and effective in preventing neurological deterioration in dogs with intraspinal spirocercosis.
Collapse
Affiliation(s)
- Nimrod Asiag
- 1Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel.,2Veterinary Specialist Referral Tipul Nimratz, Ben Shemen, Israel
| | - Orit Chai
- 2Veterinary Specialist Referral Tipul Nimratz, Ben Shemen, Israel
| | - Sapir Yodovner
- 1Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Marco Ruggeri
- 1Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Kira Rapaport
- 2Veterinary Specialist Referral Tipul Nimratz, Ben Shemen, Israel
| | - Gad Baneth
- 1Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Yaarit Nachum-Biala
- 1Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Lilach Konstantin
- 1Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | | | | | - Yaron Bruchim
- 2Veterinary Specialist Referral Tipul Nimratz, Ben Shemen, Israel
| | - Merav H Shamir
- 1Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
6
|
Hoang R, Temple C, Correia MS, Clemons J, Hendrickson RG. Characteristics of ivermectin toxicity in patients taking veterinary and human formulations for the prevention and treatment of COVID-19. Clin Toxicol (Phila) 2022; 60:1350-1355. [PMID: 36374218 DOI: 10.1080/15563650.2022.2134788] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND US poison control centers reported increased cases of ivermectin toxicity during the COVID-19 pandemic. Previous descriptions of ivermectin toxicity have evaluated heterogeneous groups with a variety of ivermectin sources and dosage patterns. We sought to compare the clinical effects of ivermectin toxicity in patients taking human- vs. veterinary-formulations and acute- vs. chronic-ingestion patterns. METHODS We performed a retrospective analysis of cases from the Oregon Poison Center of ivermectin exposures for the prevention or treatment of COVID-19 that resulted in a healthcare visit over a 24-week period (14 August 2021 - 31 January 2022). RESULTS We identified 37 cases of ivermectin toxicity. The median age of patients was 64 years, and most patients were male. The majority of patients were hospitalized (21) or treated in an emergency department (13). A minority were treated in an outpatient setting (3) and one patient died. Seventeen ingested veterinary formulations and fifteen ingested prescription tablets. Patients reported taking ivermectin for treatment (23) and prevention (14) of COVID-19. Clinical effects included neurotoxicity (30), gastrointestinal symptoms (14), and musculoskeletal complaints (7). Patients taking veterinary products took higher doses of ivermectin and had higher rates of altered mental status than those taking prescription tablets. Patients taking ivermectin chronically took smaller doses (daily dose of 13.5 mg) over a prolonged period (median 3.8 weeks) and developed toxicity that was milder than those with acute ingestions. CONCLUSION Ivermectin toxicity developed in predominantly male patients >60 years old who ingested higher than recommended doses and developed neurologic symptoms. Patients who took a veterinary formulation of ivermectin ingested large single doses or large daily doses for several days and developed rapid onset of neurotoxicity. Patients with chronic toxicity developed milder symptoms and tended to take typical therapeutic doses, but continued therapy for weeks rather than days.
Collapse
Affiliation(s)
- Ruby Hoang
- Department of Emergency Medicine, Division of Medical Toxicology, Oregon Health and Science University, Portland, OR, USA.,Oregon Poison Center, Oregon Health and Science University, Portland, OR, USA
| | - Courtney Temple
- Department of Emergency Medicine, Division of Medical Toxicology, Oregon Health and Science University, Portland, OR, USA.,Oregon Poison Center, Oregon Health and Science University, Portland, OR, USA
| | - Matthew S Correia
- Department of Emergency Medicine, Division of Medical Toxicology, Oregon Health and Science University, Portland, OR, USA.,Oregon Poison Center, Oregon Health and Science University, Portland, OR, USA
| | - Joseph Clemons
- Department of Emergency Medicine, Division of Medical Toxicology, Oregon Health and Science University, Portland, OR, USA.,Oregon Poison Center, Oregon Health and Science University, Portland, OR, USA
| | - Robert G Hendrickson
- Department of Emergency Medicine, Division of Medical Toxicology, Oregon Health and Science University, Portland, OR, USA.,Oregon Poison Center, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
7
|
Buonfrate D, Rodari P, Barda B, Page W, Einsiedel L, Watts M. Current pharmacotherapeutic strategies for Strongyloidiasis and the complications in its treatment. Expert Opin Pharmacother 2022; 23:1617-1628. [PMID: 35983698 DOI: 10.1080/14656566.2022.2114829] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Strongyloidiasis, an infection caused by the soil-transmitted helminth Strongyloides stercoralis, can lead immunocompromised people to a life-threatening syndrome. We highlight here current and emerging pharmacotherapeutic strategies for strongyloidiasis and discuss treatment protocols according to patient cohort. We searched PubMed and Embase for papers published on this topic between 1990 and May 2022. AREAS COVERED Ivermectin is the first-line drug, with an estimated efficacy of about 86% and excellent tolerability. Albendazole has a lower efficacy, with usage advised when ivermectin is not available or not recommended. Moxidectin might be a valid alternative to ivermectin, with the advantage of being a dose-independent formulation. EXPERT OPINION The standard dose of ivermectin is 200 µg/kg single dose orally, but multiple doses might be needed in immunosuppressed patients. In the case of hyperinfection, repeated doses are recommended up to 2 weeks after clearance of larvae from biological fluids, with close monitoring and further dosing based on review. Subcutaneous ivermectin is used where there is impaired intestinal absorption/paralytic ileus. In pregnant or lactating women, studies have not identified increased risk with ivermectin use. However, with limited available data, a risk-benefit assessment should be considered for each case.
Collapse
Affiliation(s)
- Dora Buonfrate
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria hospital, via Sempreboni 5, 37024 Negrar, Verona, Italy
| | - Paola Rodari
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria hospital, via Sempreboni 5, 37024 Negrar, Verona, Italy
| | - Beatrice Barda
- Ospedale La Carità, Ospedale regionale di Locarno, Locarno 6600, Switzerland
| | - Wendy Page
- Miwatj Health Aboriginal Corporation, 1424 Arnhem Rd, Nhulunbuy NT 0880, Australia
| | - Lloyd Einsiedel
- Peter Doherty Institute, University of Melbourne, 792 Elizabeth St, Melbourne VIC 3000, Australia
| | - Matthew Watts
- Centre for Infectious Diseases and Microbiology, Institute of Clinical Pathology and Medical Research - New South Wales Health Pathology and Sydney Institute for Infectious Diseases, Westmead Hospital, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
8
|
Gupta S, Vohra S, Sethi K, Gupta S, Bera BC, Kumar S, Kumar R. In vitro anti-trypanosomal effect of ivermectin on Trypanosoma evansi by targeting multiple metabolic pathways. Trop Anim Health Prod 2022; 54:240. [PMID: 35869164 PMCID: PMC9307293 DOI: 10.1007/s11250-022-03228-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 07/13/2022] [Indexed: 11/18/2022]
Abstract
High cytotoxicity and increasing resistance reports of existing chemotherapeutic agents against T. evansi have raised the demand for novel, potent, and high therapeutic index molecules for the treatment of surra in animals. In this regard, repurposing approach of drug discovery has provided an opportunity to explore the therapeutic potential of existing drugs against new organism. With this objective, the macrocyclic lactone representative, ivermectin, has been investigated for the efficacy against T. evansi in the axenic culture medium. To elucidate the potential target of ivermectin in T. evansi, mRNA expression profile of 13 important drug target genes has been studied at 12, 24, and 48 h interval. In the in vitro growth inhibition assay, ivermectin inhibited T. evansi growth and multiplication significantly (p < 0.001) with IC50 values of 13.82 μM, indicating potent trypanocidal activity. Cytotoxicity assays on equine peripheral blood mononuclear cells (PBMCs) and Vero cell line showed that ivermectin affected the viability of cells with a half-maximal cytotoxic concentration (CC50) at 17.48 and 22.05 μM, respectively. Data generated showed there was significant down-regulation of hexokinase (p < 0.001), ESAG8 (p < 0.001), aurora kinase (p < 0.001), casein kinase 1 (p < 0.001), topoisomerase II (p < 0.001), calcium ATPase 1 (p < 0.001), ribonucleotide reductase I (p < 0.05), and ornithine decarboxylase (p < 0.01). The mRNA expression of oligopeptidase B remains refractory to the exposure of the ivermectin. The arginine kinase 1 and ribonucleotide reductase II showed up-regulation on treatment with ivermectin. The ivermectin was found to affect glycolytic pathways, ATP-dependent calcium ATPase, cellular kinases, and other pathway involved in proliferation and maintenance of internal homeostasis of T. evansi. These data imply that intervention with alternate strategies like nano-formulation, nano-carriers, and nano-delivery or identification of ivermectin homologs with low cytotoxicity and high bioavailability can be explored in the future as an alternate treatment for surra in animals.
Collapse
|
9
|
Rezai MS, Ahangarkani F, Hill A, Ellis L, Mirchandani M, Davoudi A, Eslami G, Roozbeh F, Babamahmoodi F, Rouhani N, Alikhani A, Najafi N, Ghasemian R, Mehravaran H, Hajialibeig A, Navaeifar MR, Shahbaznejad L, Rahimzadeh G, Saeedi M, Alizadeh-Navai R, Moosazadeh M, Saeedi S, Razavi-Amoli SK, Rezai S, Rostami-Maskopaee F, Hosseinzadeh F, Movahedi FS, Markowitz JS, Valadan R. Non-effectiveness of Ivermectin on Inpatients and Outpatients With COVID-19; Results of Two Randomized, Double-Blinded, Placebo-Controlled Clinical Trials. Front Med (Lausanne) 2022; 9:919708. [PMID: 35783616 PMCID: PMC9244711 DOI: 10.3389/fmed.2022.919708] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/30/2022] [Indexed: 01/04/2023] Open
Abstract
Background Ivermectin which was widely considered as a potential treatment for COVID-19, showed uncertain clinical benefit in many clinical trials. Performing large-scale clinical trials to evaluate the effectiveness of this drug in the midst of the pandemic, while difficult, has been urgently needed. Methods We performed two large multicenter randomized, double-blind, placebo-controlled clinical trials evaluating the effectiveness of ivermectin in treating inpatients and outpatients with COVID-19 infection. The intervention group received ivermectin, 0.4mg/kg of body weight per day for 3 days. In the control group, placebo tablets were used for 3 days. Results Data for 609 inpatients and 549 outpatients were analyzed. In hospitalized patients, complete recovery was significantly higher in the ivermectin group (37%) compared to placebo group (28%; RR, 1.32 [95% CI, 1.04–1.66]; p-value = 0.02). On the other hand, the length of hospital stay was significantly longer in the ivermectin group with a mean of 7.98 ± 4.4 days compared to the placebo receiving group with a mean of 7.16 ± 3.2 days (RR, 0.80 [95% CI, 0.15–1.45]; p-value = 0.02). In outpatients, the mean duration of fever was significantly shorter (2.02 ± 0.11 days) in the ivermectin group versus (2.41 ± 0.13 days) placebo group with p value = 0.020. On the day seventh of treatment, fever (p-value = 0.040), cough (p-value = 0.019), and weakness (p-value = 0.002) were significantly higher in the placebo group compared to the ivermectin group. Among all outpatients, 7% in ivermectin group and 5% in placebo group needed to be hospitalized (RR, 1.36 [95% CI, 0.65–2.84]; p-value = 0.41). Also, the result of RT-PCR on day five after treatment was negative for 26% of patients in the ivermectin group versus 32% in the placebo group (RR, 0.81 [95% CI, 0.60–1.09]; p-value = 0.16). Conclusion Our data showed, ivermectin, compared with placebo, did not have a significant potential effect on clinical improvement, reduced admission in ICU, need for invasive ventilation, and death in hospitalized patients; likewise, no evidence was found to support the prescription of ivermectin on recovery, reduced hospitalization and increased negative RT-PCR assay for SARS-CoV-2 5 days after treatment in outpatients. Our findings do not support the use of ivermectin to treat mild to severe forms of COVID-19. Clinical Trial Registration www.irct.ir IRCT20111224008507N5 and IRCT20111224008507N4.
Collapse
Affiliation(s)
- Mohammad Sadegh Rezai
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
- *Correspondence: Mohammad Sadegh Rezai,
| | - Fatemeh Ahangarkani
- Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Andrew Hill
- Department of Pharmacology and Therapeutics, Liverpool University, Liverpool, United Kingdom
| | - Leah Ellis
- Faculty of Medicine, School of Public Health, Imperial College London, London, United Kingdom
| | - Manya Mirchandani
- Faculty of Medicine, School of Public Health, Imperial College London, London, United Kingdom
| | - Alireza Davoudi
- Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Gohar Eslami
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Roozbeh
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Farhang Babamahmoodi
- Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nima Rouhani
- Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Alikhani
- Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Narges Najafi
- Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Roya Ghasemian
- Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Mehravaran
- Department of Internal Medicine, Pulmonary and Critical Care Division, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Azin Hajialibeig
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Reza Navaeifar
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Leila Shahbaznejad
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Golnar Rahimzadeh
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Majid Saeedi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Alizadeh-Navai
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahmood Moosazadeh
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahab Saeedi
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Shaghayegh Rezai
- Department of Microbiology and Virology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fereshteh Rostami-Maskopaee
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Hosseinzadeh
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - John S. Markowitz
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, United States
| | - Reza Valadan
- Department of Immunology and Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
10
|
Zhang Y, Sun T, Li M, Lin Y, Liu Y, Tang S, Dai C. Ivermectin-Induced Apoptotic Cell Death in Human SH-SY5Y Cells Involves the Activation of Oxidative Stress and Mitochondrial Pathway and Akt/mTOR-Pathway-Mediated Autophagy. Antioxidants (Basel) 2022; 11:antiox11050908. [PMID: 35624772 PMCID: PMC9137967 DOI: 10.3390/antiox11050908] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 12/19/2022] Open
Abstract
Ivermectin (IVM) could cause potential neurotoxicity; however, the precise molecular mechanisms remain unclear. This study explores the cytotoxicity of IVM in human neuroblastoma (SH-SY5Y) cells and the underlying molecular mechanisms. The results show that IVM treatment (2.5–15 μM) for 24 h could induce dose-dependent cell death in SH-SY5Y cells. Compared to the control, IVM treatment significantly promoted the production of ROS, mitochondrial dysfunction, and cell apoptosis. IVM treatment also promoted mitophagy and autophagy, which were charactered by the decreased expression of phosphorylation (p)-Akt and p-mTOR proteins, increased expression of LC3II, Beclin1, ATG5, PINK, and Pakin1 proteins and autophagosome formation. N-acetylcysteine treatment significantly inhibited the IVM-induced production of ROS and cell death in SH-SY5Y cells. Autophagy inhibitor (e.g., 3-methyladenine) treatment significantly inhibited IVM-induced autophagy, oxidative stress, and cell apoptosis. Taken together, our results reveal that IVM could induce autophagy and apoptotic cell death in SH-SY5Y cells, which involved the production of ROS, activation of mitochondrial pathway, and inhibition of Akt/mTOR pathway. Autophagy inhibition improved IVM-induced oxidative stress and apoptotic cell death in SH-SY5Y cells. This current study provides new insights into understanding the molecular mechanism of IVM-induced neurotoxicity and facilitates the discovery of potential neuroprotective agents.
Collapse
Affiliation(s)
- Yuan Zhang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
| | - Tun Sun
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
| | - Meng Li
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
| | - Yanling Lin
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
| | - Yue Liu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
| | - Shusheng Tang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Correspondence: (S.T.); (C.D.)
| | - Chongshan Dai
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- Correspondence: (S.T.); (C.D.)
| |
Collapse
|
11
|
Ratain MJ, Greenblatt DJ. Drug Interactions with a Short Course of Nirmatrelvir and Ritonavir: Prescribers and Patients Beware. J Clin Pharmacol 2022; 62:925-927. [PMID: 35412678 PMCID: PMC9541177 DOI: 10.1002/jcph.2060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/07/2022] [Indexed: 11/10/2022]
Affiliation(s)
| | - David J Greenblatt
- Tufts University School of Medicine, Boston, MA, USA.,the Clinical and Translational Sciences Institute, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
12
|
Salman M, Abbas RZ, Mehmood K, Hussain R, Shah S, Faheem M, Zaheer T, Abbas A, Morales B, Aneva I, Martínez JL. Assessment of Avermectins-Induced Toxicity in Animals. Pharmaceuticals (Basel) 2022; 15:332. [PMID: 35337129 PMCID: PMC8950826 DOI: 10.3390/ph15030332] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 01/02/2023] Open
Abstract
Macrocyclic lactones, particularly the avermectins, have completely revolutionized the approaches aimed at control of parasites. These avermectins are the most widely used anti-parasitic drugs in veterinary field with sales exceeding one billion US dollars annually. However, before clinical usage, their safety evaluation in the animals is a major critical factor that must be considered. Many studies have reported the negative effects of avermectins like ivermectin, abamectin, doramectin, and eprinomectin on the host animals. These harmful effects arise from avermectins targeting GABA and glutamate-gated chloride channels present both in the parasites and the host animals. In this review, various modes of avermectins action along with the negative effects on the host like nephrotoxicity, hepatotoxicity, neurotoxicity, reproductive toxicity, and endocrine disruption were discussed in detail. Furthermore, other important issues like ecotoxicity, drug resistance, and drug residues in milk associated with avermectins usage were also discussed, which need special attention.
Collapse
Affiliation(s)
- Muhammad Salman
- Department of Parasitology, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan; (M.S.); (S.S.); (T.Z.)
| | - Rao Zahid Abbas
- Department of Parasitology, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan; (M.S.); (S.S.); (T.Z.)
| | - Khalid Mehmood
- Department of Clinical Medicine and Surgery, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan;
| | - Riaz Hussain
- Department of Pathology, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan;
| | - Sehar Shah
- Department of Parasitology, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan; (M.S.); (S.S.); (T.Z.)
| | - Mehwish Faheem
- Department of Zoology, Government College University Lahore, Lahore 54000, Pakistan;
| | - Tean Zaheer
- Department of Parasitology, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan; (M.S.); (S.S.); (T.Z.)
| | - Asghar Abbas
- Faculty of Veterinary and Animal Sciences, Muhammad Nawaz Shareef University of Agriculture Multan, Multan 59300, Pakistan;
| | - Bernardo Morales
- Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Estación Central, Santiago 9160000, Chile
| | - Ina Aneva
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - José L. Martínez
- Vicerrectoria de Investigación, Desarrollo e Innovación, Universidad de Santiago de Chile, Estación Central, Santiago 9160000, Chile
| |
Collapse
|
13
|
Kositz C, Bradley J, Hutchins H, Last A, D'Alessandro U, Marks M. Broadening the range of use cases for ivermectin - a review of the evidence. Trans R Soc Trop Med Hyg 2022; 116:201-212. [PMID: 34323283 PMCID: PMC8890779 DOI: 10.1093/trstmh/trab114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/30/2021] [Accepted: 07/13/2021] [Indexed: 11/12/2022] Open
Abstract
Ivermectin is a broad-spectrum antiparasitic agent that interferes with glutamate-gated chloride channels found in invertebrates but not in vertebrate species. Mass drug administration (MDA) with ivermectin-based regimes has been a mainstay of elimination efforts targeting onchocerciasis and lymphatic filariasis for more than 3 decades. More recently, interest in the use of ivermectin to control other neglected tropical diseases (NTDs) such as soil-transmitted helminths and scabies has grown. Interest has been further stimulated by the fact that ivermectin displays endectocidal efficacy against various Anopheles species capable of transmitting malaria. Therefore there is growing interest in using ivermectin MDA as a tool that might aid in the control of both malaria and several NTDs. In this review we outline the evidence base to date on these emerging indications for ivermectin MDA with reference to clinical and public health data and discuss the rationale for evaluating the range of impacts of a malaria ivermectin MDA on other NTDs.
Collapse
Affiliation(s)
- Christian Kositz
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - John Bradley
- MRC International Statistics and Epidemiology Group, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - Harry Hutchins
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - Anna Last
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
- Hospital for Tropical Diseases, Mortimer Market Capper Street, WC1E 6JB, London, UK
| | - Umberto D'Alessandro
- Disease Control and Elimination, Medical Research Council Unit Gambia at London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Fajara, The Gambia
| | - Michael Marks
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
- Hospital for Tropical Diseases, Mortimer Market Capper Street, WC1E 6JB, London, UK
| |
Collapse
|
14
|
Abstract
Introduction: Avermectins are common antiparasitic drugs, derived from Streptomyces bacteria that exhibit activity against arthropods and nematodes. Ivermectin, an avermectin derivative, is used as a treatment for parasitic infections in humans and domesticated animals.Discussion: Ivermectin's mechanism of action involves binding to ligand-gated ion channel receptors including glutamate, GABA, and glycine, resulting in parasitic paralysis and death. Due to varying expression of these ion channel receptors in vertebrate species, ivermectin toxicity is rarely reported in mammals. Ivermectin is also a substrate for P-glycoprotein, which limits its neurological toxicity in humans. Genetic polymorphisms in P-glycoprotein or coadministration of P-glycoprotein inhibitors may increase the neurotoxicity of ivermectin. Other toxic effects of ivermectin after therapeutic oral use include edema, rash, headache, and ocular complaints. Most of these effects are mild and short in duration. Ivermectin exhibits antiviral effects in-vitro at very high concentrations. This has led to suggestions of ivermectin as a potential treatment for SARS-CoV-2 (COVID-19) infection, although the drug's pharmacokinetic parameters reduce the likelihood that high concentrations of the drug can be achieved in-vivo.Conclusion: Due to concern for adverse events, specifically neurotoxicity, as well as a paucity of supporting evidence, the use of ivermectin as a routine treatment or preventive measure for COVID-19 infection is not recommended at this time.
Collapse
Affiliation(s)
- Kelly Johnson-Arbor
- Department of Plastic Surgery, MedStar Georgetown University Hospital, Washington, DC, USA.,National Capital Poison Center, Washington, DC, USA
| |
Collapse
|
15
|
Intravenous veterinary ivermectin in a COVID-19 patient causing neurotoxicity. IDCases 2022; 27:e01446. [PMID: 35155125 PMCID: PMC8818555 DOI: 10.1016/j.idcr.2022.e01446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 12/04/2022] Open
Abstract
Ivermectin administration for Coronavirus disease 2019 (COVID-19) infection has gained a lot of attention recently. Although ivermectin has a relatively good safety profile, serious adverse events may occur in patients given doses that are presumed experimental. Ivermectin for human use is available only as an oral formulation. Parenteral administration, as a subcutaneous injection, is possible in veterinary medicine only. In this brief report we describe an unprecedented case of a patient with severe neurotoxicity after intravenous administration of veterinary ivermectin for confirmed COVID-19 infection. The patient required hospitalization in the intensive care unit (ICU). The toxic serum concentration of ivermectin was determined by liquid chromatography/mass spectrometry - time of flight (LC/MS-TOF) with the value of 187.74 ng/mL.
Collapse
|
16
|
Temple C, Hoang R, Hendrickson RG. Toxic Effects from Ivermectin Use Associated with Prevention and Treatment of Covid-19. N Engl J Med 2021; 385:2197-2198. [PMID: 34670041 PMCID: PMC8552535 DOI: 10.1056/nejmc2114907] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
| | - Ruby Hoang
- Oregon Health and Science University, Portland, OR
| | | |
Collapse
|
17
|
Nunes B, Pinheiro D, Gomes A. Effect of sublethal concentrations of the antiparasitic ivermectin on the polychaeta species Hediste diversicolor: biochemical and behavioral responses. ECOTOXICOLOGY (LONDON, ENGLAND) 2021; 30:1841-1853. [PMID: 34224071 DOI: 10.1007/s10646-021-02444-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 06/13/2023]
Abstract
Pharmaceutical drugs have emerged as major micropollutants in aquatic ecosystems. Their presence has been systematically reported in monitoring surveys, and their wide distribution and constant presence in the wild is a direct consequence of their massive use, in both human and veterinary therapeutics. Drugs used to treat parasitic infections in livestock are major contaminants, given the amounts in which they are administered, and reach the aquatic compartment in high amounts, where they may affect non target species. Some of these drugs are prone to find their final deposit in sediments of estuarine areas, exerting their toxic effects preferentially at these locations. Sediment dwelling organisms of coastal areas, such as polychaetas, are especially prone to have their major physiological functions compromised after being exposed to pharmaceutical drugs. Ivermectin is one of the most used antiparasitic drugs, and its effects are not limited to biochemical traits, but also behavioral features may be compromised considering their neurotoxic actions. Despite these putative effects, little is known about their toxicity on polychaetas. The present study aimed to characterize the toxicity of realistic levels of ivermectin on the polychaeta Hediste diversicolor, in biochemical and behavioral terms. The obtained results showed that low levels of ivermectin are capable of causing significant disturbances in mobility and burrowing activity of exposed worms, as well as alterations of metabolic and anti-oxidant defense efficacy of exposed animals, suggesting that its environmental presence may mean a major environmental concern.
Collapse
Affiliation(s)
- B Nunes
- Departamento de Biologia da Universidade de Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal.
- Centro de Estudos do Ambiente e do Mar (CESAM), Campus de Santiago, Universidade de Aveiro, 3810-193, Aveiro, Portugal.
| | - D Pinheiro
- Departamento de Física da Universidade de Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal
| | - A Gomes
- Departamento de Biologia da Universidade de Aveiro, Campus de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
18
|
Mathachan SR, Sardana K, Khurana A. Current Use of Ivermectin in Dermatology, Tropical Medicine, and COVID-19: An Update on Pharmacology, Uses, Proven and Varied Proposed Mechanistic Action. Indian Dermatol Online J 2021; 12:500-514. [PMID: 34430453 PMCID: PMC8354388 DOI: 10.4103/idoj.idoj_298_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/16/2021] [Indexed: 12/22/2022] Open
Abstract
Ivermectin is a broad-spectrum antiparasitic drug with anti-inflammatory, anti-viral, anti-bacterial, and anti-tumor effects. In this review, we discuss the history, pharmacology, multimodal actions, indications in dermatology and tropical medicine, therapeutic and prophylactic use of ivermectin in COVID-19, safety, adverse effects, special considerations, and drug interactions of ivermectin.
Collapse
Affiliation(s)
- Sinu Rose Mathachan
- Departments of Dermatology, Venereology and Leprosy, ABVIMS and Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Kabir Sardana
- Departments of Dermatology, Venereology and Leprosy, ABVIMS and Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Ananta Khurana
- Departments of Dermatology, Venereology and Leprosy, ABVIMS and Dr. Ram Manohar Lohia Hospital, New Delhi, India
| |
Collapse
|
19
|
Lobo Y, Wheller L. A narrative review of the roles of topical permethrin and oral ivermectin in the management of infantile scabies. Australas J Dermatol 2021; 62:267-277. [PMID: 34184244 DOI: 10.1111/ajd.13654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/20/2021] [Accepted: 05/29/2021] [Indexed: 11/29/2022]
Abstract
As standard treatments are not licensed for use in the infantile population, the treatment of scabies in this age group can be challenging. We review the relevant evidence to determine the roles of topical permethrin and oral ivermectin in the management of infantile scabies. Demographic and clinical data were collected from relevant English articles published from January 2000 to December 2020. Complete resolution was observed in 100% of infants younger than two months treated with permethrin, and 87.6% of infants aged 12 months or less and/or children weighing under 15 kg treated with ivermectin. Adverse effects from permethrin use were limited to local eczematous reactions. Adverse effects from ivermectin use included mildly elevated creatine kinase levels, eczema flare-ups, diarrhoea, vomiting, irritability, pruritus and pustular skin reactions. Overall, both permethrin and ivermectin appear to have an acceptable safety profile in infants. Permethrin is highly effective as a first-line therapy for scabies in infants younger than two months. Ivermectin use is recommended when authorised topical treatment has failed, in crusted scabies, in cases where compliance with topical agents may be problematic, and in infants with severely inflamed or broken skin where prescription of topical therapies would likely cause cutaneous and systemic toxicity. Additional high-quality studies are needed to guide best practice in the management of infantile scabies.
Collapse
Affiliation(s)
- Yolanka Lobo
- Department of Dermatology, Mater Misericordiae Health Services, Brisbane, Queensland, Australia
| | - Laura Wheller
- Department of Dermatology, Mater Misericordiae Health Services, Brisbane, Queensland, Australia.,Department of Dermatology, Queensland Children's Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
20
|
A systematic review and an individual patient data meta-analysis of ivermectin use in children weighing less than fifteen kilograms: Is it time to reconsider the current contraindication? PLoS Negl Trop Dis 2021; 15:e0009144. [PMID: 33730099 PMCID: PMC7968658 DOI: 10.1371/journal.pntd.0009144] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/13/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Oral ivermectin is a safe broad spectrum anthelminthic used for treating several neglected tropical diseases (NTDs). Currently, ivermectin use is contraindicated in children weighing less than 15 kg, restricting access to this drug for the treatment of NTDs. Here we provide an updated systematic review of the literature and we conducted an individual-level patient data (IPD) meta-analysis describing the safety of ivermectin in children weighing less than 15 kg. METHODOLOGY/PRINCIPAL FINDINGS A systematic review was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) for IPD guidelines by searching MEDLINE via PubMed, Web of Science, Ovid Embase, LILACS, Cochrane Database of Systematic Reviews, TOXLINE for all clinical trials, case series, case reports, and database entries for reports on the use of ivermectin in children weighing less than 15 kg that were published between 1 January 1980 to 25 October 2019. The protocol was registered in the International Prospective Register of Systematic Reviews (PROSPERO): CRD42017056515. A total of 3,730 publications were identified, 97 were selected for potential inclusion, but only 17 sources describing 15 studies met the minimum criteria which consisted of known weights of children less than 15 kg linked to possible adverse events, and provided comprehensive IPD. A total of 1,088 children weighing less than 15 kg were administered oral ivermectin for one of the following indications: scabies, mass drug administration for scabies control, crusted scabies, cutaneous larva migrans, myiasis, pthiriasis, strongyloidiasis, trichuriasis, and parasitic disease of unknown origin. Overall a total of 1.4% (15/1,088) of children experienced 18 adverse events all of which were mild and self-limiting. No serious adverse events were reported. CONCLUSIONS/SIGNIFICANCE Existing limited data suggest that oral ivermectin in children weighing less than 15 kilograms is safe. Data from well-designed clinical trials are needed to provide further assurance.
Collapse
|
21
|
Metabolism and interactions of Ivermectin with human cytochrome P450 enzymes and drug transporters, possible adverse and toxic effects. Arch Toxicol 2021; 95:1535-1546. [PMID: 33719007 PMCID: PMC7956433 DOI: 10.1007/s00204-021-03025-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/09/2021] [Indexed: 12/23/2022]
Abstract
The review presents metabolic properties of Ivermectin (IVM) as substrate and inhibitor of human P450 (P450, CYP) enzymes and drug transporters. IVM is metabolized, both in vivo and in vitro, by C-hydroxylation and O-demethylation reactions catalyzed by P450 3A4 as the major enzyme, with a contribution of P450 3A5 and 2C9. In samples from both in vitro and in vivo metabolism, a number of metabolites were detected and as major identified metabolites were 3″-O-demethylated, C4-methyl hydroxylated, C25 isobutyl-/isopropyl-hydroxylated, and products of oxidation reactions. Ivermectin inhibited P450 2C9, 2C19, 2D6, and CYP3A4 with IC50 values ranging from 5.3 μM to no inhibition suggesting that it is no or weak inhibitor of the enzymes. It is suggested that P-gp (MDR1) transporter participate in IVM efflux at low drug concentration with a slow transport rate. At the higher, micromolar concentration range, which saturates MDR1 (P-gp), MRP1, and to a lesser extent, MRP2 and MRP3 participate in IVM transport across physiological barriers. IVM exerts a potent inhibition of P-gp (ABCB1), MRP1 (ABCC1), MRP2 (ABCC2), and BCRP1 (ABCG2), and medium to weak inhibition of OATP1B1 (SLC21A6) and OATP1B3 (SLCOB3) transport activity. The metabolic and transport properties of IVM indicate that when IVM is co-administered with other drugs/chemicals that are potent inhibitors/inducers P4503A4 enzyme and of MDR1 (P-gp), BCRP or MRP transporters, or when polymorphisms of the drug transporters and P450 3A4 exist, drug–drug or drug–toxic chemical interactions might result in suboptimal response to the therapy or to toxic effects.
Collapse
|
22
|
Fricke-Galindo I, Falfán-Valencia R. Pharmacogenetics Approach for the Improvement of COVID-19 Treatment. Viruses 2021; 13:413. [PMID: 33807592 PMCID: PMC7998786 DOI: 10.3390/v13030413] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/22/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
The treatment of coronavirus disease 2019 (COVID-19) has been a challenge. The efficacy of several drugs has been evaluated and variability in drug response has been observed. Pharmacogenetics could explain this variation and improve patients' outcomes with this complex disease; nevertheless, several disease-related issues must be carefully reviewed in the pharmacogenetic study of COVID-19 treatment. We aimed to describe the pharmacogenetic variants reported for drugs used for COVID-19 treatment (remdesivir, oseltamivir, lopinavir, ritonavir, azithromycin, chloroquine, hydroxychloroquine, ivermectin, and dexamethasone). In addition, other factors relevant to the design of pharmacogenetic studies were mentioned. Variants in CYP3A4, CYP3A5, CYP2C8, CY2D6, ABCB1, ABCC2, and SLCO1B1, among other variants, could be included in pharmacogenetic studies of COVID-19 treatment. Besides, nongenetic factors such as drug-drug interactions and inflammation should be considered in the search for personalized therapy of COVID-19.
Collapse
Affiliation(s)
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| |
Collapse
|
23
|
Manchanda K, Singh J, Bhagat R, Tiwana IK, Singh H. Safety of pharmacological options for the management of COVID-19 in pregnant women: An Indian perspective. INTERNATIONAL JOURNAL OF RISK & SAFETY IN MEDICINE 2021; 32:3-17. [PMID: 33386817 DOI: 10.3233/jrs-200060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a viral illness caused by severe acute respiratory syndrome coronavirus 2 (SARS- CoV-2) presenting with pulmonary and extra-pulmonary manifestations. The first case was reported in Wuhan, China in December 2019 and it has rapidly progressed to the form of a pandemic. The presentation is mild in about 80 percent of the cases but the disease can also progress to a severe form of respiratory illness leading to acute respiratory distress syndrome (ARDS) and sometimes multi-organ failure, especially in people with other co-morbidities. Pregnant women also appear to be at a greater risk of acquiring a severe infection due to physiological changes during pregnancy. Many drugs with in vitro activity against the virus or an immunomodulatory effect have been considered for repurposing or have been tried as off-label drugs. The safety data regarding the use of newly approved or off-label or investigational drugs in pregnant women is limited and this poses a great challenge for clinicians. Therefore, it is important to know the utility and safety of the medications to avoid untoward adverse effects on pregnant women and fetuses. In this review, we aim to provide an overview of the approved, off-label, unlicensed, new and some promising pharmacological options for their use in the treatment of COVID-19 and the safety profile in pregnancy in an Indian scenario.
Collapse
Affiliation(s)
- Kavita Manchanda
- Fellow in Reproductive Medicine, Milann Fertility Centre, Bangalore, India
| | - Jasbir Singh
- Associate Professor, Department of Pharmacology, Government Medical College and Rajindra Hospital, Patiala, India
| | - Ranjeev Bhagat
- Assistant Professor, Department of Pathology, Government Medical College and Hospital, Chandigarh, India
| | - Ilmjot Kaur Tiwana
- MBBS Intern, Government Medical College and Rajindra Hospital, Patiala, India
| | - Harmanjit Singh
- Assistant Professor, Department of Pharmacology, Government Medical College and Hospital, Chandigarh, India
| |
Collapse
|
24
|
Udaykumar P, Shetty B, Kundapur A. Drug interactions of ivermectin with a focus on COVID-19 treatment. MULLER JOURNAL OF MEDICAL SCIENCES AND RESEARCH 2021. [DOI: 10.4103/mjmsr.mjmsr_19_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
25
|
Mudassar F, Shen H, O'Neill G, Hau E. Targeting tumor hypoxia and mitochondrial metabolism with anti-parasitic drugs to improve radiation response in high-grade gliomas. J Exp Clin Cancer Res 2020; 39:208. [PMID: 33028364 PMCID: PMC7542384 DOI: 10.1186/s13046-020-01724-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
High-grade gliomas (HGGs), including glioblastoma and diffuse intrinsic pontine glioma, are amongst the most fatal brain tumors. These tumors are associated with a dismal prognosis with a median survival of less than 15 months. Radiotherapy has been the mainstay of treatment of HGGs for decades; however, pronounced radioresistance is the major obstacle towards the successful radiotherapy treatment. Herein, tumor hypoxia is identified as a significant contributor to the radioresistance of HGGs as oxygenation is critical for the effectiveness of radiotherapy. Hypoxia plays a fundamental role in the aggressive and resistant phenotype of all solid tumors, including HGGs, by upregulating hypoxia-inducible factors (HIFs) which stimulate vital enzymes responsible for cancer survival under hypoxic stress. Since current attempts to target tumor hypoxia focus on reducing oxygen demand of tumor cells by decreasing oxygen consumption rate (OCR), an attractive strategy to achieve this is by inhibiting mitochondrial oxidative phosphorylation, as it could decrease OCR, and increase oxygenation, and could therefore improve the radiation response in HGGs. This approach would also help in eradicating the radioresistant glioma stem cells (GSCs) as these predominantly rely on mitochondrial metabolism for survival. Here, we highlight the potential for repurposing anti-parasitic drugs to abolish tumor hypoxia and induce apoptosis of GSCs. Current literature provides compelling evidence that these drugs (atovaquone, ivermectin, proguanil, mefloquine, and quinacrine) could be effective against cancers by mechanisms including inhibition of mitochondrial metabolism and tumor hypoxia and inducing DNA damage. Therefore, combining these drugs with radiotherapy could potentially enhance the radiosensitivity of HGGs. The reported efficacy of these agents against glioblastomas and their ability to penetrate the blood-brain barrier provides further support towards promising results and clinical translation of these agents for HGGs treatment.
Collapse
Affiliation(s)
- Faiqa Mudassar
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, Westmead Institute for Medical Research, NSW, Westmead, Australia
| | - Han Shen
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, Westmead Institute for Medical Research, NSW, Westmead, Australia.
- Sydney Medical School, University of Sydney, NSW, Sydney, Australia.
| | - Geraldine O'Neill
- Children's Cancer Research Unit, The Children's Hospital at Westmead, NSW, Westmead, Australia
- Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, NSW, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, NSW, Sydney, Australia
| | - Eric Hau
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, Westmead Institute for Medical Research, NSW, Westmead, Australia
- Sydney Medical School, University of Sydney, NSW, Sydney, Australia
- Department of Radiation Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, NSW, Westmead, Australia
- Blacktown Hematology and Cancer Centre, Blacktown Hospital, NSW, Blacktown, Australia
| |
Collapse
|
26
|
Omolo CA, Soni N, Fasiku VO, Mackraj I, Govender T. Update on therapeutic approaches and emerging therapies for SARS-CoV-2 virus. Eur J Pharmacol 2020; 883:173348. [PMID: 32634438 PMCID: PMC7334944 DOI: 10.1016/j.ejphar.2020.173348] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 02/09/2023]
Abstract
The global pandemic of coronavirus disease 2019 (COVID-19), caused by novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in over 7,273,958 cases with almost over 413,372 deaths worldwide as per the WHO situational report 143 on COVID-19. There are no known treatment regimens with proven efficacy and vaccines thus far, posing an unprecedented challenge to identify effective drugs and vaccines for prevention and treatment. The urgency for its prevention and cure has resulted in an increased number of proposed treatment options. The high rate and volume of emerging clinical trials on therapies for COVID-19 need to be compared and evaluated to provide scientific evidence for effective medical options. Other emerging non-conventional drug discovery techniques such as bioinformatics and cheminformatics, structure-based drug design, network-based methods for prediction of drug-target interactions, artificial intelligence (AI) and machine learning (ML) and phage technique could provide alternative routes to discovering potent Anti-SARS-CoV2 drugs. While drugs are being repurposed and discovered for COVID-19, novel drug delivery systems will be paramount for efficient delivery and avoidance of possible drug resistance. This review describes the proposed drug targets for therapy, and outcomes of clinical trials that have been reported. It also identifies the adopted treatment modalities that are showing promise, and those that have failed as drug candidates. It further highlights various emerging therapies and future strategies for the treatment of COVID-19 and delivery of Anti-SARS-CoV2 drugs.
Collapse
Affiliation(s)
- Calvin A Omolo
- United States International University-Africa, School of Pharmacy and Health Sciences, Department of Pharmaceutics, P. O. Box 14634-00800, Nairobi, Kenya; Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
| | - Nikki Soni
- United States International University-Africa, School of Pharmacy and Health Sciences, Department of Pharmaceutics, P. O. Box 14634-00800, Nairobi, Kenya
| | - Victoria Oluwaseun Fasiku
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Irene Mackraj
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Thirumala Govender
- United States International University-Africa, School of Pharmacy and Health Sciences, Department of Pharmaceutics, P. O. Box 14634-00800, Nairobi, Kenya.
| |
Collapse
|
27
|
Safety of oral ivermectin during pregnancy: a systematic review and meta-analysis. LANCET GLOBAL HEALTH 2020; 8:e92-e100. [PMID: 31839144 PMCID: PMC7613514 DOI: 10.1016/s2214-109x(19)30453-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/23/2019] [Accepted: 10/11/2019] [Indexed: 12/11/2022]
Abstract
Background About 3·7 billion doses of ivermectin have been distributed in mass drug administration (MDA) campaigns globally over the past 30 years. At 10−100 times higher than current human doses, ivermectin is a known teratogen in mammals. During these campaigns with recommended doses, pregnant women might be inadvertently exposed. We therefore aimed to evaluate the existing evidence for serious and non-serious adverse events after ivermectin exposure in pregnant women. Methods For this systematic review and meta-analysis, we searched relevant databases and trial registry platforms on July 15, 2018, for randomised controlled trials (RCTs) and observational studies that reported adverse events in pregnant women. We did not use language or date restrictions. Outcomes of interest were spontaneous abortions, stillbirths, congenital anomalies, and neonatal death (serious adverse events), as well as maternal morbidity, preterm births, and low birthweight (adverse events). The risk of bias was assessed using the Newcastle-Ottawa Scale for observational studies and the Cochrane Risk of Bias Tool for RCTs. We did the meta-analysis of observational studies and RCTs separately. The quality of evidence was assessed using the GRADE approach. The study protocol is registered with PROSPERO, protocol CRD42016046914. Findings We identified 147 records, of which only five observational studies and one RCT were included for quantitative analysis; these studies were published between 1990 and 2008, and were done in six African countries. 893 women with 899 pregancy outcomes were included, of whom 496 pregnant women (500 pregnancy outcomes) received ivermectin inadvertently during MDA campaigns in the observational studies and 397 pregnant women (399 pregnancy outcomes) purposely received ivermectin as part of the open-label RCT. No study reported neonatal deaths, maternal morbidity, preterm births, or low birthweight. It is unclear whether exposure to ivermectin during pregnancy increases the risk of spontaneous abortions and stillbirths (odds ratio [OR] 1·15 [95% CI 0·75−1·78] with very low certainty of evidence for the four observational studies and 0·62 [0·18−2·14] with very low certainty of evidence for the RCT) or congenital anomalies (OR 1·69 [95% CI 0·83−3·41] with very low certainty of evidence for the five observational studies and 1·10 [0·07−17·65] with very low certainty of evidence for the RCT). Interpretation There is insufficient evidence to conclude on the safety profile of ivermectin during pregnancy. Treatment campaigns should focus additional efforts on preventing inadvertent treatment of pregnant women. Funding Unitaid.
Collapse
|
28
|
Billingsley P, Binka F, Chaccour C, Foy B, Gold S, Gonzalez-Silva M, Jacobson J, Jagoe G, Jones C, Kachur P, Kobylinski K, Last A, Lavery JV, Mabey D, Mboera D, Mbogo C, Mendez-Lopez A, Rabinovich NR, Rees S, Richards F, Rist C, Rockwood J, Ruiz-Castillo P, Sattabongkot J, Saute F, Slater H, Steer A, Xia K, Zullinger R. A Roadmap for the Development of Ivermectin as a Complementary Malaria Vector Control Tool. Am J Trop Med Hyg 2020; 102:3-24. [PMID: 31971144 PMCID: PMC7008306 DOI: 10.4269/ajtmh.19-0620] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/22/2019] [Indexed: 12/14/2022] Open
Abstract
In the context of stalling progress against malaria, resistance of mosquitoes to insecticides, and residual transmission, mass drug administration (MDA) of ivermectin, an endectocide used for neglected tropical diseases (NTDs), has emerged as a promising complementary vector control method. Ivermectin reduces the life span of Anopheles mosquitoes that feed on treated humans and/or livestock, potentially decreasing malaria parasite transmission when administered at the community level. Following the publication by WHO of the preferred product characteristics for endectocides as vector control tools, this roadmap provides a comprehensive view of processes needed to make ivermectin available as a vector control tool by 2024 with a completely novel mechanism of action. The roadmap covers various aspects, which include 1) the definition of optimal dosage/regimens for ivermectin MDA in both humans and livestock, 2) the risk of resistance to the drug and environmental impact, 3) ethical issues, 4) political and community engagement, 5) translation of evidence into policy, and 6) operational aspects of large-scale deployment of the drug, all in the context of a drug given as a prevention tool acting at the community level. The roadmap reflects the insights of a multidisciplinary group of global health experts who worked together to elucidate the path to inclusion of ivermectin in the toolbox against malaria, to address residual transmission, counteract insecticide resistance, and contribute to the end of this deadly disease.
Collapse
Affiliation(s)
| | - Fred Binka
- University of Health and Allied Sciences
| | | | | | | | | | | | | | | | | | | | - Anna Last
- London School of Hygiene and Tropical Medicine
| | | | - David Mabey
- London School of Hygiene and Tropical Medicine
| | | | | | | | | | | | | | - Cassidy Rist
- Virginia-Maryland College of Veterinary Medicine at Virginia Tech
| | | | | | | | | | | | | | - Kang Xia
- School of Plant and Environmental Sciences, Virginia Tech
| | - Rose Zullinger
- US President’s Malaria Initiative/US Centers for Disease Control and Prevention
| |
Collapse
|
29
|
Liu J, Zhang K, Cheng L, Zhu H, Xu T. Progress in Understanding the Molecular Mechanisms Underlying the Antitumour Effects of Ivermectin. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:285-296. [PMID: 32021111 PMCID: PMC6982461 DOI: 10.2147/dddt.s237393] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022]
Abstract
Ivermectin, a dihydro derivative of avermectin (AVM), was introduced into the veterinary, agricultural and aquaculture markets for animal health in 1981. Ivermectin was soon adopted in 1987 as a human medicine that was originally used for the treatment of onchocerciasis, a parasitic infection. Since then, ivermectin has also been used to control other human diseases and has exerted a significant effect on human health and welfare. In the past decade, many published studies have attempted to determine the role of ivermectin in cancer. In this review, we summarize the published studies to define the current progress in the characterization of ivermectin. Ivermectin causes cell death in cancer cell lines by inducing PAK1-mediated cytostatic autophagy, caspase-dependent apoptosis and immunogenic cell death (ICD) through the modulation of some pathways, including the WNT-T cell factor (TCF), Hippo and Akt/mTOR pathways. Ivermectin can affect the growth and proliferation of cancer cells and plays several different roles, such as its functions as an RNA helicase, a small-molecule mimetic of the surface-induced dissociation (SID) peptide, an activator of chloride channel receptors, and an inducer of mitochondrial dysfunction and oxidative stress. In addition, ivermectin induces the multidrug resistance protein (MDR), has potent anti-mitotic activity, targets angiogenesis and inhibits cancer stem-like cells (CSCs). Many studies have proven that ivermectin exerts antitumour effects and might thus benefit patients with cancer after sufficient clinical trials.
Collapse
Affiliation(s)
- Jian Liu
- Department of Obstetrics and Gynecology, Jilin University Second Hospital, ChangChun 130041, People's Republic of China
| | - Kun Zhang
- Department of Obstetrics and Gynecology, Jilin University Second Hospital, ChangChun 130041, People's Republic of China
| | - Lin Cheng
- Department of Obstetrics and Gynecology, Jilin University Second Hospital, ChangChun 130041, People's Republic of China
| | - He Zhu
- Department of Obstetrics and Gynecology, Jilin University Second Hospital, ChangChun 130041, People's Republic of China
| | - Tianmin Xu
- Department of Obstetrics and Gynecology, Jilin University Second Hospital, ChangChun 130041, People's Republic of China
| |
Collapse
|
30
|
Gowtham S, Karthikeyan K. Wonder drug for worms: A review of three decades of ivermectin use in dermatology. Indian J Dermatol Venereol Leprol 2019; 85:674-678. [PMID: 31552839 DOI: 10.4103/ijdvl.ijdvl_840_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Saravanan Gowtham
- Department of Dermatology Venereology and Leprosy, Sri Manakula Vinayagar Medical College and Hospital, Puducherry, India
| | - Kaliaperumal Karthikeyan
- Department of Dermatology Venereology and Leprosy, Sri Manakula Vinayagar Medical College and Hospital, Puducherry, India
| |
Collapse
|
31
|
Trexler AW, Knudsen GA, Nicklisch SCT, Birnbaum LS, Cannon RE. 2,4,6-Tribromophenol Exposure Decreases P-Glycoprotein Transport at the Blood-Brain Barrier. Toxicol Sci 2019; 171:463-472. [PMID: 31368499 PMCID: PMC6760274 DOI: 10.1093/toxsci/kfz155] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/12/2019] [Accepted: 07/09/2019] [Indexed: 01/24/2023] Open
Abstract
2,4,6-Tribromophenol (TBP, CAS No. 118-79-6) is a brominated chemical used in the production of flame-retardant epoxy resins and as a wood preservative. In marine environments, TBP is incorporated into shellfish and consumed by predatory fish. Food processing and water treatment facilities produce TBP as a byproduct. 2,4,6-Tribromophenol has been detected in human blood and breast milk. Biologically, TBP interferes with estrogen and thyroid hormone signaling, which regulate important transporters of the blood-brain barrier (BBB). The BBB is a selectively permeable barrier characterized by brain microvessels which are composed of endothelial cells mortared by tight-junction proteins. ATP-binding cassette (ABC) efflux transporters on the luminal membrane facilitate the removal of unwanted endobiotics and xenobiotics from the brain. In this study, we examined the in vivo and ex vivo effects of TBP on two important transporters of the BBB: P-glycoprotein (P-gp, ABCB1) and Multidrug Resistance-associated Protein 2 (MRP2, ABCC2), using male and female rats and mice. 2,4,6-Tribromophenol exposure ex vivo resulted in a time- (1-3 h) and dose- (1-100 nM) dependent decrease in P-gp transport activity. MRP2 transport activity was unchanged under identical conditions. Immunofluorescence and western blotting measured decreases in P-gp expression after TBP treatment. ATPase assays indicate that TBP is not a substrate and does not directly interact with P-gp. In vivo dosing with TBP (0.4 µmol/kg) produced decreases in P-gp transport. Co-treatment with selective protein kinase C (PKC) inhibitors prevented the TBP-mediated decreases in P-gp transport activity.
Collapse
Affiliation(s)
- Andrew W Trexler
- NCI Laboratory of Toxicology and Toxicokinetics, Research Triangl Park, North Carolina, 27709
| | - Gabriel A Knudsen
- NCI Laboratory of Toxicology and Toxicokinetics, Research Triangl Park, North Carolina, 27709
| | - Sascha C T Nicklisch
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California San Diego, La Jolla 92093, California
- Department of Environmental Toxicology, University of California Davis 95616, Davis, California
| | - Linda S Birnbaum
- NCI Laboratory of Toxicology and Toxicokinetics, Research Triangl Park, North Carolina, 27709
| | - Ronald E Cannon
- NCI Laboratory of Toxicology and Toxicokinetics, Research Triangl Park, North Carolina, 27709
| |
Collapse
|
32
|
Jiang L, Wang P, Sun YJ, Wu YJ. Ivermectin reverses the drug resistance in cancer cells through EGFR/ERK/Akt/NF-κB pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:265. [PMID: 31215501 PMCID: PMC6580523 DOI: 10.1186/s13046-019-1251-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/28/2019] [Indexed: 12/23/2022]
Abstract
Background Discovery and development of novel drugs that are capable of overcoming drug resistance in tumor cells are urgently needed clinically. In this study, we sought to explore whether ivermectin (IVM), a macrolide antiparasitic agent, could overcome the resistance of cancer cells to the therapeutic drugs. Methods We used two solid tumor cell lines (HCT-8 colorectal cancer cells and MCF-7 breast cancer cells) and one hematologic tumor cell line (K562 chronic myeloid leukemia cells), which are resistant to the chemotherapeutic drugs vincristine and adriamycin respectively, and two xenograft mice models, including the solid tumor model in nude mice with the resistant HCT-8 cells and the leukemia model in NOD/SCID mice with the resistant K562 cells to investigate the reversal effect of IVM on the resistance in vitro and in vivo. MTT assay was used to investigate the effect of IVM on cancer cells growth in vitro. Flow cytometry, immunohistochemistry, and immunofluorescence were performed to investigate the reversal effect of IVM in vivo. Western blotting, qPCR, luciferase reporter assay and ChIP assay were used to detect the molecular mechanism of the reversal effect. Octet RED96 system and Co-IP were used to determine the interactions between IVM and EGFR. Results Our results indicated that ivermectin at its very low dose, which did not induce obvious cytotoxicity, drastically reversed the resistance of the tumor cells to the chemotherapeutic drugs both in vitro and in vivo. Mechanistically, ivermectin reversed the resistance mainly by reducing the expression of P-glycoprotein (P-gp) via inhibiting the epidermal growth factor receptor (EGFR), not by directly inhibiting P-gp activity. Ivermectin bound with the extracellular domain of EGFR, which inhibited the activation of EGFR and its downstream signaling cascade ERK/Akt/NF-κB. The inhibition of the transcriptional factor NF-κB led to the reduced P-gp transcription. Conclusions These findings demonstrated that ivermectin significantly enhanced the anti-cancer efficacy of chemotherapeutic drugs to tumor cells, especially in the drug-resistant cells. Thus, ivermectin, a FDA-approved antiparasitic drug, could potentially be used in combination with chemotherapeutic agents to treat cancers and in particular, the drug-resistant cancers. Electronic supplementary material The online version of this article (10.1186/s13046-019-1251-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lu Jiang
- Laboratory of Molecular Toxicology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1-5 Beichenxilu Rd., Chaoyang, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Pan Wang
- Laboratory of Molecular Toxicology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1-5 Beichenxilu Rd., Chaoyang, Beijing, 100101, China
| | - Ying-Jian Sun
- Laboratory of Molecular Toxicology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1-5 Beichenxilu Rd., Chaoyang, Beijing, 100101, China. .,Department of Veterinary Medicine, Beijing University of Agriculture, Beinonglu Rd, Changping, Beijing, 102206, China.
| | - Yi-Jun Wu
- Laboratory of Molecular Toxicology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1-5 Beichenxilu Rd., Chaoyang, Beijing, 100101, China.
| |
Collapse
|
33
|
Konecny P, Weatherall CJ, Adhikari S, Duflou J, Marjoniemi V, Pretorius CJ, McWhinney B. Case Report: Subcutaneous Ivermectin Pharmacokinetics in Disseminated Strongyloides Infection: Plasma and Postmortem Analysis. Am J Trop Med Hyg 2018; 99:1580-1582. [PMID: 30334520 DOI: 10.4269/ajtmh.18-0387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Parenteral ivermectin treatment of disseminated strongyloidiasis and hyperinfection is increasing, although not licensed in humans and with limited pharmacokinetic data available. Plasma and postmortem tissue analysis in an human immunodeficiency virus (HIV)/hepatitis C virus-positive man with disseminated strongyloidiasis suggests loading subcutaneous ivermectin doses are required, from which the central nervous system is protected.
Collapse
Affiliation(s)
- Pamela Konecny
- University of New South Wales, Sydney, Australia.,St. George Hospital, Sydney, Australia
| | | | - Suman Adhikari
- University of New South Wales, Sydney, Australia.,St. George Hospital, Sydney, Australia
| | - Johan Duflou
- University of Sydney, Sydney, Australia.,Forensic Medicine Associates P/L, Sydney, Australia
| | - Veli Marjoniemi
- University of New South Wales, Sydney, Australia.,St. George Hospital, Sydney, Australia
| | - Carel J Pretorius
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia.,Pathology Queensland, Brisbane, Australia
| | | |
Collapse
|
34
|
Chen C, Lee MH, Weng CF, Leong MK. Theoretical Prediction of the Complex P-Glycoprotein Substrate Efflux Based on the Novel Hierarchical Support Vector Regression Scheme. Molecules 2018; 23:E1820. [PMID: 30037151 PMCID: PMC6100076 DOI: 10.3390/molecules23071820] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/13/2022] Open
Abstract
P-glycoprotein (P-gp), a membrane-bound transporter, can eliminate xenobiotics by transporting them out of the cells or blood⁻brain barrier (BBB) at the expense of ATP hydrolysis. Thus, P-gp mediated efflux plays a pivotal role in altering the absorption and disposition of a wide range of substrates. Nevertheless, the mechanism of P-gp substrate efflux is rather complex since it can take place through active transport and passive permeability in addition to multiple P-gp substrate binding sites. A nonlinear quantitative structure⁻activity relationship (QSAR) model was developed in this study using the novel machine learning-based hierarchical support vector regression (HSVR) scheme to explore the perplexing relationships between descriptors and efflux ratio. The predictions by HSVR were found to be in good agreement with the observed values for the molecules in the training set (n = 50, r² = 0.96, qCV2 = 0.94, RMSE = 0.10, s = 0.10) and test set (n = 13, q² = 0.80⁻0.87, RMSE = 0.21, s = 0.22). When subjected to a variety of statistical validations, the developed HSVR model consistently met the most stringent criteria. A mock test also asserted the predictivity of HSVR. Consequently, this HSVR model can be adopted to facilitate drug discovery and development.
Collapse
Affiliation(s)
- Chun Chen
- Department of Chemistry, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| | - Ming-Han Lee
- Department of Chemistry, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| | - Ching-Feng Weng
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| | - Max K Leong
- Department of Chemistry, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Shoufeng, Hualien 97401, Taiwan.
| |
Collapse
|
35
|
Dail MB, Meek EC, Chambers HW, Chambers JE. In vitro P-glycoprotein activity does not completely explain in vivo efficacy of novel centrally effective oxime acetylcholinesterase reactivators. Drug Chem Toxicol 2018; 42:403-408. [PMID: 29722548 DOI: 10.1080/01480545.2018.1461902] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Novel-substituted phenoxyalkyl pyridinium oxime acetylcholinesterase (AChE) reactivators (US patent 9,227,937) that showed convincing evidence of penetration into the brains of intact rats were developed by our laboratories. The oximes separated into three groups based on their levels of brain AChE reactivation following exposure of rats to the sarin surrogate nitrophenyl isopropyl methylphosphonate (NIMP). P-glycoprotein (P-gp) is a major blood-brain barrier (BBB) transporter and requires ATP for efflux. To determine if P-gp affinity screening could be used to reduce animal use, we measured in vitro oxime-stimulated ATPase activity to see if the in vivo reactivation efficacies related to the oximes' functions as P-gp substrates. High efficacy oximes were expected to be poor P-gp substrates, thus remaining in the brain longer. The high efficacy oximes (24-35% brain AChE reactivation) were worse P-gp substrates than the low efficacy oximes (0-7% brain AChE reactivation). However, the oxime group with medium in vivo reactivation of 10-17% were even worse P-gp substrates than the high efficacy group so their reactivation ability was not reflected by P-gp export. The results suggest that in vitro P-gp ATPase activity can remove the low efficacy oximes from in vivo testing, but is not sufficient to differentiate between the top two tiers.
Collapse
Affiliation(s)
- Mary Beth Dail
- a Center for Environmental Health Sciences, Department of Basic Sciences , College of Veterinary Medicine, Mississippi State University, Mississippi State , MS , USA
| | - Edward Caldwell Meek
- a Center for Environmental Health Sciences, Department of Basic Sciences , College of Veterinary Medicine, Mississippi State University, Mississippi State , MS , USA
| | - Howard Wayne Chambers
- b Department of Biochemistry, Molecular Biology, Entomology, and Plant Pathology , Entomology Unit, Mississippi State University, Mississippi State , MS , USA
| | - Janice Elaine Chambers
- a Center for Environmental Health Sciences, Department of Basic Sciences , College of Veterinary Medicine, Mississippi State University, Mississippi State , MS , USA
| |
Collapse
|
36
|
Wilkins AL, Steer AC, Cranswick N, Gwee A. Question 1: Is it safe to use ivermectin in children less than five years of age and weighing less than 15 kg? Arch Dis Child 2018; 103:514-519. [PMID: 29463522 DOI: 10.1136/archdischild-2017-314505] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/11/2018] [Accepted: 01/15/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Amanda L Wilkins
- Department of General Medicine, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| | - Andrew C Steer
- Department of General Medicine, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Noel Cranswick
- Department of General Medicine, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Amanda Gwee
- Department of General Medicine, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| |
Collapse
|
37
|
Lee JS, Suh JW, Kim ES, Lee HG. Preparation and Characterization of Mucoadhesive Nanoparticles for Enhancing Cellular Uptake of Coenzyme Q10. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:8930-8937. [PMID: 28933847 DOI: 10.1021/acs.jafc.7b03300] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The mucoadhesive nanoparticles (NPs) for oral delivery of coenzyme Q10 (CoQ10) were prepared using natural mucoadhesive polysaccharides, chitosan (CS), and dextran sulfate sodium salt (DS) in order to improve the solubility, cellular uptake, and thermo- and photostability of CoQ10. CoQ10-loaded NPs were prepared in the range of 340-450 nm with an entrapment efficiency of 60-98%. The mucoadhesiveness and cellular uptake of NPs were evaluated by measuring the amount of mucin adsorbed on NPs and CoQ10 absorbed in Caco-2 cells, respectively. CS/DS NPs had higher mucoadhesive strength than CS/sodium triphosphate pentabasic NPs (control group). Moreover, the solubility, cellular uptake, thermo- and photostability of CS/DS NPs were significantly improved compared with non-nanoencapsulated free CoQ10. Particularly, CS/DS NPs prepared with 0.5 mg/mL of CS and DS produced the highest mucoadhesiveness, solubility, cellular uptake, and cellular antioxidant activity. Thus, mucoadhesive CS/DS NPs may be an effective oral delivery platform for improving bioavailability of CoQ10.
Collapse
Affiliation(s)
- Ji-Soo Lee
- Department of Food and Nutrition, Hanyang University , 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| | - Ji Woon Suh
- Department of Food and Nutrition, Hanyang University , 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| | - Eun Suh Kim
- Department of Food and Nutrition, Hanyang University , 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| | - Hyeon Gyu Lee
- Department of Food and Nutrition, Hanyang University , 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| |
Collapse
|
38
|
Colebunders R, Mandro M, Mukendi D, Dolo H, Suykerbuyk P, Van Oijen M. Ivermectin Treatment in Patients With Onchocerciasis-Associated Epilepsy: Protocol of a Randomized Clinical Trial. JMIR Res Protoc 2017; 6:e137. [PMID: 28855148 PMCID: PMC5597797 DOI: 10.2196/resprot.7186] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/10/2017] [Accepted: 05/25/2017] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Many studies have reported an association between epilepsy, nodding syndrome (NS), and onchocerciasis (river blindness). A high prevalence of epilepsy has been noted particularly in onchocerciasis hyperendemic areas where onchocerciasis is not or insufficiently controlled with mass ivermectin distribution. There is evidence that increasing the coverage of ivermectin reduces the incidence of epilepsy, and anecdotal evidence suggests a reduction in seizure frequency in onchocerciasis-associated epilepsy (OAE) patients who receive ivermectin. Finding an alternative treatment for epilepsy in these patients will have major consequences. OBJECTIVE The goal of the study is to assess whether ivermectin treatment decreases the frequency of seizures and leads to seizure freedom in OAE patients, including patients with NS. If we are able to demonstrate such an effect, this would strengthen the argument that onchocerciasis is causing epilepsy and therefore we should increase our efforts to eliminate onchocerciasis. METHODS We will conduct a randomized clinical trial in the Democratic Republic of Congo to compare seizure freedom in onchocerciasis-infested epilepsy patients who receive immediate ivermectin treatment with delayed (after 4 months) ivermectin treatment. All participants will simultaneously receive antiepilepsy drugs (AEDs) according to local guidelines for epilepsy treatment. The primary endpoint is seizure freedom defined as no seizures during the 4 month of follow-up. Secondary endpoint is significant (>50%) seizure reduction compared to baseline seizure frequency. Reduction of seizures will be compared between ivermectin and nonivermectin arms. RESULTS Start of enrollment is planned for August 2017, and we expect to have enrolled all 110 participants by December 2017. Results are expected in June 2018. CONCLUSIONS If ivermectin treatment in addition to AEDs is able to lead to seizure freedom or significantly reduces seizure frequency in OAE patients, this will have major consequences for epilepsy treatment in onchocerciasis-endemic regions. Ivermectin is donated for free and in non Loa-Loa-endemic regions has negligible side effects. Reducing the burden of epilepsy will have a major impact on quality of life and socioeconomic status of families with affected members in Africa. TRIAL REGISTRATION ClinicalTrials.gov NCT03052998; https://clinicaltrials.gov/ct2/show/NCT03052998 (Archived by WebCite at http://www.webcitation.org/6roFVQSG0).
Collapse
Affiliation(s)
| | - Michel Mandro
- Provincial Ministry of Health, Bunia, The Democratic Republic Of The Congo
| | - Deby Mukendi
- Centre Neuro Psycho Pathologique, University of Kinshasa, Kinshasa, The Democratic Republic Of The Congo
| | - Housseini Dolo
- University of Antwerp, Global Health Institute, Antwerp, Belgium
| | | | | |
Collapse
|
39
|
Cytochrome P450/ABC transporter inhibition simultaneously enhances ivermectin pharmacokinetics in the mammal host and pharmacodynamics in Anopheles gambiae. Sci Rep 2017; 7:8535. [PMID: 28819225 PMCID: PMC5561046 DOI: 10.1038/s41598-017-08906-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/14/2017] [Indexed: 01/06/2023] Open
Abstract
Mass administration of endectocides, drugs that kill blood-feeding arthropods, has been proposed as a complementary strategy to reduce malaria transmission. Ivermectin is one of the leading candidates given its excellent safety profile. Here we provide proof that the effect of ivermectin can be boosted at two different levels by drugs inhibiting the cytochrome or ABC transporter in the mammal host and the target mosquitoes. Using a mini-pig model, we show that drug-mediated cytochrome P450/ABC transporter inhibition results in a 3-fold increase in the time ivermectin remains above mosquito-killing concentrations. In contrast, P450/ABC transporter induction with rifampicin markedly impaired ivermectin absorption. The same ketoconazole-mediated cytochrome/ABC transporter inhibition also occurs outside the mammal host and enhances the mortality of Anopheles gambiae. This was proven by using the samples from the mini-pig experiments to conduct an ex-vivo synergistic bioassay by membrane-feeding Anopheles mosquitoes. Inhibiting the same cytochrome/xenobiotic pump complex in two different organisms to simultaneously boost the pharmacokinetic and pharmacodynamic activity of a drug is a novel concept that could be applied to other systems. Although the lack of a dose-response effect in the synergistic bioassay warrants further exploration, our study may have broad implications for the control of parasitic and vector-borne diseases.
Collapse
|
40
|
Kigen G, Edwards G. Drug-transporter mediated interactions between anthelminthic and antiretroviral drugs across the Caco-2 cell monolayers. BMC Pharmacol Toxicol 2017; 18:20. [PMID: 28468637 PMCID: PMC5415745 DOI: 10.1186/s40360-017-0129-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 02/28/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Drug interactions between antiretroviral drugs (ARVs) and anthelminthic drugs, ivermectin (IVM) and praziquantel (PZQ) were assessed by investigating their permeation through the Caco-2 cell monolayers in a transwell. The impact of anthelminthics on the transport of ARVs was determined by assessing the apical to basolateral (AP → BL) [passive] and basolateral to apical (BL → AP) [efflux] directions alone, and in presence of an anthelminthic. The reverse was conducted for the assessment of the influence of ARVs on anthelminthics. METHODS Samples from the AP and BL compartments were taken at 60, 120, 180 and 240 min and quantified either by HPLC or radiolabeled assay using a liquid scintillating counter for the respective drugs. Transepithelial resistance (TEER) was used to assess the integrity of the monolayers. The amount of compound transported per second (apparent permeability, Papp) was calculated for both AP to BL (PappAtoB), and BL to AP (PappBtoA) movements. Samples collected after 60 min were used to determine the efflux ratio (ER), quotient of secretory permeability and absorptive permeability (PappBL-AP/PappAP-BL). The reverse, (PappAP-BL/PappBL-AP) constituted the uptake ratio. The impact of SQV, EFV and NVP on the transport of both IVM and PZQ were investigated. The effect of LPV on the transport of IVM was also determined. The influence of IVM on the transport of SQV, NVP, LPV and EFV; as well as the effect PZQ on the transport of SQV of was also investigated, and a two-tailed p value of <0.05 was considered significant. RESULTS IVM significantly inhibited the efflux transport (BL → AP movement) of LPV (ER; 6.7 vs. 0.8, p = 0.0038) and SQV (ER; 3.1 vs. 1.2 p = 0.00328); and increased the efflux transport of EFV (ER; 0.7 vs. 0.9, p = 0.031) suggesting the possibility of drug transporter mediated interactions between the two drugs. NVP increased the efflux transport of IVM (ER; 0.8 vs. 1.8, p = 0.0094). CONCLUSIONS The study provides in vitro evidence of potential interactions between IVM, an anthelminthic drug with antiretroviral drugs; LPV, SQV, NVP and EFV. Further investigations should be conducted to investigate the possibility of in vivo interactions.
Collapse
Affiliation(s)
- Gabriel Kigen
- Department of Pharmacology and Toxicology, Moi University School of Medicine, P.O. Box 4606, 30100 Eldoret, Kenya
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE UK
| | - Geoffrey Edwards
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE UK
| |
Collapse
|
41
|
Chaccour C, Hammann F, Rabinovich NR. Ivermectin to reduce malaria transmission I. Pharmacokinetic and pharmacodynamic considerations regarding efficacy and safety. Malar J 2017; 16:161. [PMID: 28434401 PMCID: PMC5402169 DOI: 10.1186/s12936-017-1801-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
Ivermectin is an endectocide that has been used broadly in single dose community campaigns for the control of onchocerciasis and lymphatic filariasis for more than 30 years. There is now interest in the potential use of ivermectin regimens to reduce malaria transmission, envisaged as community-wide campaigns tailored to transmission patterns and as complement of the local vector control programme. The development of new ivermectin regimens or other novel endectocides will require integrated development of the drug in the context of traditional entomological tools and endpoints. This document examines the main pharmacokinetic and pharmacodynamic parameters of the medicine and their potential influence on its vector control efficacy and safety at population level. This information could be valuable for trial design and clinical development into regulatory and policy pathways.
Collapse
Affiliation(s)
- Carlos Chaccour
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain. .,Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique. .,Instituto de Salud Tropical Universidad de Navarra, Pamplona, Spain.
| | - Felix Hammann
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, Basel, Switzerland
| | - N Regina Rabinovich
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain.,Harvard T.H. Chan School of Public Health, Boston, USA
| |
Collapse
|
42
|
Huynh N, Arabian N, Naito A, Louie S, Jakowec MW, Asatryan L, Davies DL. Preclinical development of moxidectin as a novel therapeutic for alcohol use disorder. Neuropharmacology 2016; 113:60-70. [PMID: 27641072 DOI: 10.1016/j.neuropharm.2016.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/14/2016] [Accepted: 09/15/2016] [Indexed: 02/08/2023]
Abstract
Current pharmacotherapies for alcohol used disorder (AUD) are few and relatively ineffective illustrating the need for the development of new, effective medications. Using a translational approach, our laboratory reported that ivermectin, an FDA-approved, human and animal anti-parasitic agent, can significantly reduce ethanol intake in male and female mice across different drinking paradigms. Extending this line of investigation, the current paper investigated the utility of moxidectin (MOX), an analogue of ivermectin, to reduce ethanol intake. Notably, MOX is widely held to have lower neurotoxicity potential and improved margin of safety compared to ivermectin. Using a 24-h-two-bottle choice paradigm, MOX significantly reduced ethanol intake in a dose dependent manner in both male and female C57BL/6J mice, respectively (1.25-7.5 mg/kg) and (1.25-10 mg/kg). Further, multi-day administration of MOX (2.5 mg/kg; intraperitoneal injection) for 5 consecutive days significantly reduced ethanol intake in both the 24-h-two-bottle choice and Drinking-in-the-Dark paradigms in female mice. No overt signs of behavioral toxicity were observed. Notably in both male and female mice, MOX significantly reduced ethanol intake starting approximately 4 h post-injection. Using a Xenopus oocyte expression system, we found that MOX significantly potentiated P2X4 receptor (P2X4R) function and antagonized the inhibitory effects of ethanol on ATP-gated currents in P2X4Rs. This latter finding represents the first report of MOX having activity on P2X4Rs. In addition, MOX potentiated GABAA receptors, but to a lesser degree as compared to ivermectin supporting the hypothesis that MOX would be advantageous (compared to ivermectin) with respect to reducing contraindications. Overall, the results illustrate the potential for development of MOX as a novel pharmacotherapy for the treatment of AUD.
Collapse
Affiliation(s)
- Nhat Huynh
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Natalie Arabian
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Anna Naito
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Stan Louie
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Michael W Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, 1333 San Pablo Street, Los Angeles, CA 90033, USA
| | - Liana Asatryan
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Daryl L Davies
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA.
| |
Collapse
|
43
|
Mohapatra S, Sahoo AJ. Drug-Induced Psychosis Associated with Albendazole-Ivermectin Combination Therapy in a 10-Year-Old Child. J Child Adolesc Psychopharmacol 2015; 25:817-8. [PMID: 26683000 DOI: 10.1089/cap.2015.0143] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
| | - Alok Jyoti Sahoo
- Mental Health Institute, SBC Medical College , Cuttack, Orissa, India
| |
Collapse
|
44
|
Lacher SE, Skagen K, Veit J, Dalton R, Woodahl EL. P-Glycoprotein Transport of Neurotoxic Pesticides. J Pharmacol Exp Ther 2015; 355:99-107. [PMID: 26272936 DOI: 10.1124/jpet.115.226373] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/12/2015] [Indexed: 01/03/2023] Open
Abstract
P-glycoprotein (P-gp) has been associated with a number of neurodegenerative diseases, including Parkinson's disease, although the mechanisms remain unclear. Altered transport of neurotoxic pesticides has been proposed in Parkinson's disease, but it is unknown whether these pesticides are P-gp substrates. We used three in vitro transport models, stimulation of ATPase activity, xenobiotic-induced cytotoxicity, and inhibition of rhodamine-123 efflux, to evaluate P-gp transport of diazinon, dieldrin, endosulfan, ivermectin, maneb, 1-methyl-4-phenyl-4-phenylpyridinium ion (MPP(+)), and rotenone. Diazinon and rotenone stimulated ATPase activity in P-gp-expressing membranes, with Vmax values of 22.4 ± 2.1 and 16.8 ± 1.0 nmol inorganic phosphate/min per mg protein, respectively, and Km values of 9.72 ± 3.91 and 1.62 ± 0.51 µM, respectively, compared with the P-gp substrate verapamil, with a Vmax of 20.8 ± 0.7 nmol inorganic phosphate/min per mg protein and Km of 0.871 ± 0.172 μM. None of the other pesticides stimulated ATPase activity. We observed an increased resistance to MPP(+) and rotenone in LLC-MDR1-WT cells compared with LLC-vector cells, with 15.4- and 2.2-fold increases in EC50 values, respectively. The resistance was reversed in the presence of the P-gp inhibitor verapamil. None of the other pesticides displayed differential cytotoxicity. Ivermectin was the only pesticide to inhibit P-gp transport of rhodamine-123, with an IC50 of 0.249 ± 0.048 μM. Our data demonstrate that dieldrin, endosulfan, and maneb are not P-gp substrates or inhibitors. We identified diazinon, MPP(+), and rotenone as P-gp substrates, although further investigation is needed to understand the role of P-gp transport in their disposition in vivo and associations with Parkinson's disease.
Collapse
Affiliation(s)
- Sarah E Lacher
- Department of Biomedical and Pharmaceutical Sciences (S.E.L., K.S., J.V., R.D., E.L.W.), Center for Environmental Health Sciences (S.E.L.), and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (E.L.W.)
| | - Kasse Skagen
- Department of Biomedical and Pharmaceutical Sciences (S.E.L., K.S., J.V., R.D., E.L.W.), Center for Environmental Health Sciences (S.E.L.), and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (E.L.W.)
| | - Joachim Veit
- Department of Biomedical and Pharmaceutical Sciences (S.E.L., K.S., J.V., R.D., E.L.W.), Center for Environmental Health Sciences (S.E.L.), and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (E.L.W.)
| | - Rachel Dalton
- Department of Biomedical and Pharmaceutical Sciences (S.E.L., K.S., J.V., R.D., E.L.W.), Center for Environmental Health Sciences (S.E.L.), and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (E.L.W.)
| | - Erica L Woodahl
- Department of Biomedical and Pharmaceutical Sciences (S.E.L., K.S., J.V., R.D., E.L.W.), Center for Environmental Health Sciences (S.E.L.), and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (E.L.W.)
| |
Collapse
|
45
|
Dunstand-Guzmán E, Peña-Chora G, Hallal-Calleros C, Pérez-Martínez M, Hernández-Velazquez VM, Morales-Montor J, Flores-Pérez FI. Acaricidal effect and histological damage induced by Bacillus thuringiensis protein extracts on the mite Psoroptes cuniculi. Parasit Vectors 2015; 8:285. [PMID: 26003809 PMCID: PMC4451748 DOI: 10.1186/s13071-015-0890-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 05/07/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The mite Psoroptes cuniculi is a common worldwide ectoparasite and the most frequently found in rabbit farms. It causes significant economic losses on commercial rabbit breeding associated with poor leather quality, reduced conception rates, weight loss, poor growth and death. Several strategies have been proposed for the treatment of mange caused by this mite, ranging from the use of acaricides, entomopathogenic fungi, essential oils and vaccines. However, therapy and control of both human scabies and animal mange are still based mainly on the use of drugs and chemicals such as ivermectin, which involves disadvantages including genotoxic and cytotoxic effects, resistance and environmental damage. Bacillus thuringiensis is a bacterium, innocuous for human being, domestic animals and plants that produces highly biodegradable proteins, and has been used worldwide for biological control. The aim of this work was to find an alternative treatment based on biological control for scabies caused by Psoroptes cuniculi, using protein extracts from strains of Bacillus thuringiensis. METHODS P. cuniculi mites were obtained from naturally infected New Zealand rabbits, and different doses of protein from B. thuringiensis were added to the mites. We measured mortality and obtained the median lethal concentration and median lethal times. For histological analysis, the mites were fixed in 10% formalin, processed according to the paraffin embedded tissue technique. Sections were stained with hematoxylin-eosin to observe the general histological structure. RESULTS We report here for the first time evidence about the in vitro acaricidal effect caused by the strain GP532 of B. thuringiensis on the mite Psoroptes cuniculi, with an LC50 of 1.3 mg/ml and a LT50 of 68 h. Histological alterations caused by B. thuringiensis on this mite, included the presence of dilated intercellular spaces in the basal membrane, membrane detachment of the peritrophic matrix and morphological alterations in columnar cells of the intestine. CONCLUSIONS Since this mite is an obligate ectoparasite that affects rabbits, goats, horses, cows and sheep, B. thuringiensis protein extracts are proposed as a potential treatment for biological control of mange in farm animals.
Collapse
Affiliation(s)
- Emmanuel Dunstand-Guzmán
- Facultad de Ciencias Agropecuarias, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, 62209, Cuernavaca, Morelos, Mexico.
| | - Guadalupe Peña-Chora
- Centro de Investigaciones Biológicas, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, 62209, Cuernavaca, Morelos, Mexico.
| | - Claudia Hallal-Calleros
- Facultad de Ciencias Agropecuarias, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, 62209, Cuernavaca, Morelos, Mexico.
| | - Mario Pérez-Martínez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Av. Universidad 3000, Col. Copilco, 04510, Ciudad de México, Mexico.
| | - Víctor Manuel Hernández-Velazquez
- Centro de Investigación en Biotecnología, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, 62209, Cuernavaca, Morelos, Mexico.
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, DF 04510, México, Mexico.
| | - Fernando Iván Flores-Pérez
- Facultad de Ciencias Agropecuarias, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, 62209, Cuernavaca, Morelos, Mexico.
| |
Collapse
|
46
|
Bates N, Chatterton J, Robbins C, Wells K, Hughes J, Stone M, Campbell A. Lipid infusion in the management of poisoning: a report of 6 canine cases. VETERINARY RECORD CASE REPORTS 2013. [DOI: 10.1136/vetreccr.101036rep] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- N. Bates
- Veterinary Poisons Information ServiceMedical Toxicology and Information ServicesMary Sheridan HouseLondonSE1 9RYUK
| | - J. Chatterton
- Grove Veterinary Centre2 Hibbert Street, New MillsHigh PeakDerbyshireSK22 3JJUK
| | - C. Robbins
- Castle Vets Pet Healthcare Centre1 Tilehurst RoadReadingRG1 7TWUK
| | - K. Wells
- Kingfisher Veterinary PracticeSouth Road Vet CentreMountfield RoadTauntonSomersetTA1 3BQUK
| | - J. Hughes
- Blake Veterinary GroupShowground RoadBridgwaterSomersetTA6 6AJUK
| | - M. Stone
- RSPCA Bristol & District Branch48 Albert RoadSt PhilipsBristolBS2 0XAUK
| | - A. Campbell
- Veterinary Poisons Information ServiceMedical Toxicology and Information ServicesMary Sheridan HouseLondonSE1 9RYUK
| |
Collapse
|
47
|
Bhalla D, Dumas M, Preux PM. Neurological manifestations of filarial infections. HANDBOOK OF CLINICAL NEUROLOGY 2013; 114:235-42. [PMID: 23829914 DOI: 10.1016/b978-0-444-53490-3.00018-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Filarial infections cause a huge public health burden wherever they are endemic. These filaria may locate anywhere in the human body. Their manifestations and pathogenic mechanisms, except the most common ones, are rarely investigated systematically. Their neurological manifestations, however, are being increasingly recognized particularly with onchocerciasis or Loa loa infections, Wuchereria bancrofti, or Mansonella perstans. The risk of developing these manifestations may also increase in cases that harbor multiple filariasis or coinfections, for instance as with Plasmodium. The microfilaria of Onchocerca and Loa loa are seen in cerebrospinal fluid. The pathogenesis of neurological manifestations of these infections is complex; however, pathogenic reactions may be caused by mechanical disruption, e.g., degeneration often followed by granulomas, causing fibrosis or mass effects on other tissues, vascular lesions, e.g., vascular block of cerebral vessels, or disordered inflammatory responses resulting in meningitis, encephalitis or localized inflammatory responses. The chances of having neurological manifestations may also depend upon the frequency and"heaviness"of infection over a lifetime. Hence, this type of infection should no longer be considered a disease of the commonly affected areas but one that may produce systemic effects or other manifestations, and these should be considered in populations where they are endemic.
Collapse
Affiliation(s)
- Devender Bhalla
- INSERM UMR 1094, Tropical Neuroepidemiology, Limoges, France; University of Limoges, School of Medicine, Institute of Neuroepidemiology and Tropical Neurology, CNRS FR 3503 GEIST, Limoges, France; Centre Hospitalier Universitaire, Limoges, France
| | | | | |
Collapse
|
48
|
Abstract
Scabies prevalence remains unacceptably high in many regions throughout the world. Infestation with scabies significantly impacts quality of life and is linked to pyoderma and consequently to severe long-term sequelae such as post-streptococcal glomerulonephritis. In the past, control programs using topical treatments have met with poor compliance; however, the highly effective oral agent ivermectin may offer a new paradigm in scabies management. Problems still exist with insensitive diagnostic tests, questions concerning mite reservoirs, and restrictions on who can receive ivermectin. Despite these difficulties, the elimination of scabies in communities worst affected may soon be possible.
Collapse
|
49
|
Spencer PS, Vandemaele K, Richer M, Palmer VS, Chungong S, Anker M, Ayana Y, Opoka ML, Klaucke BN, Quarello A, Tumwine JK. Nodding syndrome in Mundri county, South Sudan: environmental, nutritional and infectious factors. Afr Health Sci 2013; 13:183-204. [PMID: 24235915 PMCID: PMC3824482 DOI: 10.4314/ahs.v13i2.2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Nodding Syndrome is a seizure disorder of children in Mundri County, Western Equatoria, South Sudan. The disorder is reported to be spreading in South Sudan and northern Uganda. OBJECTIVE To describe environmental, nutritional, infectious, and other factors that existed before and during the de novo 1991 appearance and subsequent increase in cases through 2001. METHODS Household surveys, informant interviews, and case-control studies conducted in Lui town and Amadi village in 2001-2002 were supplemented in 2012 by informant interviews in Lui and Juba, South Sudan. RESULTS Nodding Syndrome was associated with Onchocerca volvulus and Mansonella perstans infections, with food use of a variety of sorghum (serena) introduced as part of an emergency relief program, and was inversely associated with a history of measles infection. There was no evidence to suggest exposure to a manmade neurotoxic pollutant or chemical agent, other than chemically dressed seed intended for planting but used for food. Food use of cyanogenic plants was documented, and exposure to fungal contaminants could not be excluded. CONCLUSION Nodding Syndrome in South Sudan has an unknown etiology. Further research is recommended on the association of Nodding Syndrome with onchocerciasis/mansonelliasis and neurotoxins in plant materials used for food.
Collapse
Affiliation(s)
- P S Spencer
- Department of Neurology, School of Medicine; Senior Scientist, Center for Research on Occupational and Environmental Toxicology, and Director, Global Health Center, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, L356, Portland, Oregon 97239, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hamdani N, Doukhan R, Picard A, TAmouza R, Leboyer M. A bipolar disorder patient becoming asymptomatic after adjunctive anti-filiarasis treatment: a case report. BMC Psychiatry 2013; 13:81. [PMID: 23497411 PMCID: PMC3623878 DOI: 10.1186/1471-244x-13-81] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 02/26/2013] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Evidence suggests that neurotropic infectious agents might be involved in bipolar disorder. So far, few have been written for the association between parasitic infection and bipolar disorder. Filariasis is a parasitic disease acting ruthlessly via mosquitos and affecting more than 120 million people worldwide. We present here, to our knowledge, the first description of a filariasis infected manic bipolar disorder patient fully improved in terms of psychiatric symptoms by anti-heminthic treatment. CASE PRESENTATION The patient is a 31 years-old man native of Congo. At inclusion, he presented a severe manic episode with dangerous behaviour unresolved by classic treatments. A diagnosis of filariasis bancrofti infection was made after the discovery of a systemic hypereosinophilia. Therefore, a bi-therapy of anthelmintics was conducted allowing a successful improvement with clear reduction of agitation and aggressive behaviours that could not be attributed to a modification of psychotropic treatments or filarial encephalopathy or acute disseminated encephalomyelitis. CONCLUSION The ineffectiveness of psychotropic treatment of a manic episode requires the evaluation of co-morbid medical conditions such as infections which can interfere with adequate mood stabilizing medication. Filariasis by inducing chronic inflammation and immunopathologic reactions seems to play a major role in infected affective disorders patients by changing levels of cytokines of the Th1 system or indirectly damaging the brain tissue. The beneficial combination of antihelmintics and mood stabilizers, in this case, could be explained by the potential of such association to downregulate neuroinflammation and excitotoxicity processes.Altogether, these data pinpoint the requirement to explore the parasitic infectious status in case of bipolar disorder patients resistant to classic treatments and originating or living in endemic geographical areas.
Collapse
Affiliation(s)
- Nora Hamdani
- Faculté de médecine, Université Paris Est, Créteil 94000, France.
| | - Raphaël Doukhan
- A. Chenevier, Pôle de psychiatrie, AP-HP, Hopital H. Mondor, Créteil 94000, France
| | - Aline Picard
- A. Chenevier, Pôle de psychiatrie, AP-HP, Hopital H. Mondor, Créteil 94000, France
| | - Ryad TAmouza
- AP-HP, Laboratoire Jean Dausset d’Immunologie et d’Histocompatibilité & INSERM, UMRS 940, Hôpital Saint Louis, Paris, France,Univ Paris Diderot, Paris, F75010, France
| | - Marion Leboyer
- Faculté de médecine, Université Paris Est, Créteil 94000, France,A. Chenevier, Pôle de psychiatrie, AP-HP, Hopital H. Mondor, Créteil 94000, France,Equipe Psychiatrie Génétique, INSERM, U955, Créteil, 94000, France,Fondation FondaMental, Créteil, 94000, France
| |
Collapse
|