The largest database of trusted experimental protocols

Amodiaquine

Amodiaquine is a 4-aminoquinoline antimalarial drug used for the treatment and prevention of malaria caused by Plasmodium falciparum.
It is effective against chloroqiune-resistant strains of the parasite.
Amodiaquine has a similar chemical structure to chloroquine but exhibits a longer half-life and different metabolic profile.
The mechanism of action involves interference with heme detoxification within the parasite's digestive vacuole.
Despite its efficacy, amodiaqine usage has been limited due to safety concerns, including the risk of liver toxicity and agranlucytosis with prolonged treatment.
Reasearch protocols for amodiaqine must be carefully optimized to ensure reproducibility and accuracy of results.

Most cited protocols related to «Amodiaquine»

The WWARN QA/QC proficiency testing program for pharmacology laboratories assesses the ability of pharmacology laboratories to assay blood or plasma samples for concentrations of antimalarial compounds and their metabolites. Participation in the proficiency testing program is open to all laboratories doing either therapeutic efficacy studies or other research on antimalarial drug exposure. The program currently offers plasma-based samples for eight antimalarial drug compounds and metabolites: chloroquine/desethylchloroquine, mefloquine/carboxymefloquine, primaquine/carboxyprimaquine, amodiaquine/desethylamodiaquine, piperaquine, lumefantrine/desbutyl-lumefantrine, dihydroartemisinin, and artesunate. Commercially obtained and controlled plasma is spiked with accurately weighed certified reference materials. All active ingredients and the plasma are controlled by the manufacturer and reflected in certificates of analysis. Each analyte is sent in a range of concentrations, including the highest and lowest concentrations expected to be found in clinical samples (Table 1), which allows each laboratory to test the limits of its assay.
Full text: Click here
Publication 2014
Amodiaquine Antimalarials artenimol Artesunate Biological Assay BLOOD carboxymefloquine carboxyprimaquine Chloroquine desethylamodiaquine desethylchloroquine Drug Compounding Lumefantrine Mefloquine piperaquine Plasma Primaquine Therapeutics

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2011
Amodiaquine Biological Assay Cells chloroquine phosphate Cytostatic Agents Ethanol Fluorescence Medical Devices Mefloquine Hydrochloride Parasite Control Parasitemia Parasites Pellets, Drug Pets Pharmaceutical Preparations Primaquine Diphosphate Protoplasm Quinidine Quinine SYBR Green I Technique, Dilution Verapamil Hydrochloride Volumes, Packed Erythrocyte
Enrollment was initiated in January 2007 in the Saponé health district in the Bazèga province where the Centre National de Recherche et de Formation sur le Paludisme (CNRFP) has maintained a continuous demographic surveillance system (DSS) since 2005. The Sapone DSS is located in the centre of Burkina Faso, 40 km at the south from the capital, Ouagadougou in the Sudan-Sahelian eco-climatic zone (isohyets 600–800). The DSS covers an area of 1700 Km2 within a plateau dissected by the Nazinon river (White Volta river). Mean temperature recorded during 2007 was 29°C and total rainfall for the same period was 713 mm. The climate is characterized by a rainy season from June to September and dry season from October to May. The total resident population is estimated as 100,000 inhabitants and infant and children mortality is 105.3 per 1000 live births. Children below five years represent 19% of the population and total fertility rate is an estimated 45‰. The predominating ethnic groups in the area are Mossi and Fulani. Most of the population depends on subsistence farming of millet and domestic animals (poultry, cattle etc.). Houses are typically made of mud walls and grass or corrugated iron roofs [9] . One reference district hospital (CMA de Saponé with 32 beds) and 14 peripheral health facilities constitute the government health network in the district.
Study randomization was performed at two levels to achieve target sample sizes of PCD and ACD cohorts: i) two of the 14 peripheral health centres catchment areas composed of 13 villages were randomly selected, ii) 9 villages of the 13 villages were randomized to conduct passive case detection and 4 to conduct active case detection of malaria over one year follow-up. All 13 villages were situated within ≈7.5 km of the peripheral health centres, facilitating access to each PCD surveillance system.
Malaria transmission in the region is markedly seasonal and intense during the rainy season. The entomological inoculation rate is estimated at 0.3 and 44.4 infective bites/person/month during the dry and rainy seasons, respectively [10] (link). The main malaria vectors are Anopheles gambiae, An. Arabiensis and An funestus. P falciparum accounts for a 90% of malaria infections. Oral chloroquine was still in use in the communities during the study period despite parasite resistance of 57% (Sirima and al. unpublished data). The first line treatment for uncomplicated malaria in Burkina is now Coartem® or Artesunate Amodiaquine. In the study region these medications are distributed primarily by peripheral health centres; there is no private clinic or pharmacy alternative although prohibited or sub-standard antimalarials are sold from the local market. In 2007, bed net coverage was less than 5% of the DSS population, reflecting the absence of large scale bed net distribution in the region.
Full text: Click here
Publication 2013
Amodiaquine Animals, Domestic Anopheles gambiae Antimalarials Artesunate Bites Cattle Child Chloroquine Climate Cloning Vectors Coartem Ethnic Groups Fowls, Domestic Infant Infection Iron Malaria Millets Parasites Pharmaceutical Preparations Poaceae Rain Rivers SELL protein, human Transmission, Communicable Disease Vaccination
In a previous analysis of the samples collected 24 hours apart, we found that not all clones present in a host were detected within a single sample. Twenty-one percent of all msp1F3 alleles and 28% of all MS16 clones were missed on a single day [30] (link). Thus, we used the combined genotyping data from both day 1 and day 2, except for samples from enrolment and final visits, where only 1 venous blood sample was taken.
The force of new P. vivax blood-stage infections (molFOB) was generated by counting the number of genotypes in each interval that had not been present in the preceding interval. An 8 to 9 weeks interval started on the first day after a regular cross-sectional visit and included all samples collected during passive case detection over two months plus the samples collected at the end of the interval. molFOB was also determined for both markers combined, msp1F3 and MS16. In case of discrepancy between the markers for an 8-weeks interval the higher estimate from either marker was used. This approach corrected for imperfect resolution and detectability of a single marker.
Genotyping cannot directly identify relapses; molFOB measures the combination of primary blood-stage infections and those caused by relapsing hypnozoites. Thus, homologous relapses occurring in two subsequent 2-month intervals would be misclassified as persisting clones. New Guinean P. vivax strains are known to relapse rapidly [31] (link), however in regions of high transmission, relapsing clones are usually of a different genotype than the initial blood stage infection [24] (link). As a consequence the number of homologous relapses that were not detected is expected to be relatively small.
In line with the pharmacokinetic properties of the drugs [32] (link)–[34] (link), children were not considered at risk for two weeks after treatment with artemether-lumefantrine and four weeks after treatment with amodiaquine (AQ) plus sulphadoxine-pyramethamine (SP). The force of blood-stage infection for each child and interval was subsequently converted into the number of new clones acquired per year-at-risk.
Similar to previous analyses of P. falciparummolFOI [23] (link), generalized linear mixed models (GLMMs) were used for analyses of force of blood-stage infection as well as for incidence of P. vivax episodes. These models were chosen because they allowed the fixed effects to be specified separately from the random effects (i.e. repeated measurements from the same child over time and unmeasured village factors). Furthermore, the random-effects model allowed for decomposition of the error into between-village and within-village variation.
We fit a Poisson GLMM model with a log link to relate the fixed and (Gaussian) random effects to the number of clinical episodes experienced during a two month interval (defined as febrile illness plus P. vivax >500 parasites/µl). Covariates were selected based on earlier analyses of the same data [27] (link). Seasonality was characterized by two readily interpretable parameters: the amplitude, which was half the range between the peak and trough, and the phase, which was the location of the first zero crossing in a cycle relative to the origin in time (in this case, the first week of the year). For computational convenience, they were replaced by sine and cosine terms with fixed phases. For all outcomes except prevalence, an offset was fit to adjust for years at risk. Estimation of these models was done using the LME4 package in R version 2.12 [35] . All point estimates provided throughout the text (except those for seasonal effects) were obtained from cubic splines fit using generalized additive models (Figure 1) using the MGCV package in R version 2.12 [36] . For a more detailed description of the statistical approaches see [23] (link). Point estimates for seasonal peaks and troughs were obtained from the GLMMs by setting all other values of the covariates at their means. For the analyses of the effect of exposure on the relationship between age and incidence of P. vivax malaria, children were stratified into terciles according to the average molFOB during the entire follow-up.
Full text: Click here
Publication 2013
Aftercare Alleles Amodiaquine BLOOD Child Clone Cells Cuboid Bone Fever Genotype Hematologic Tests Infection Lumefantrine, Artemether Malaria, Vivax Parasites Pharmaceutical Preparations Relapse Short Interspersed Nucleotide Elements Strains Sulfadoxine Transmission, Communicable Disease Vasculitis Veins
The study was carried out in Dielmo, a village situated in a Sudan-savannah region of central Senegal, on the marshy bank of a small permanent stream, where anopheline mosquitoes breed all year round [22] (link). Malaria transmission is intense and perennial, with a mean 258 and 132 infected bites per person per year during 1990–2006 and 2007–2010, respectively [21] (link). From 1990 to 2010, we did a longitudinal study involving most of the population of the village (all 247 inhabitants of the village in June 1990 at the beginning of the project, 468 of 509 inhabitants in December 2010).
The study area, the procedures of medical, parasitological, entomological and epidemiological surveillance and the main characteristics of malaria in this village have been described previously [21] (link), [22] (link). Briefly, during 20 years, we visited all households daily, and collected nominative information on the presence or absence in the village of each individual we had enrolled, their location when absent, and the presence of fever or other symptoms. We systematically recorded body temperature at home three times a week (every second day except Sunday) in children younger than 5 years, and in older children and adults in case of suspected fever or fever-related symptoms. In cases of fever or other symptoms, blood testing was done by finger prick at our dispensary located in the village, and we provided detailed medical examination and specific treatment. The dispensary created for our project was open 24 h a day, 7 days a week, to allow both active and passive case detection. To investigate asymptomatic malaria carriage, we performed cross-sectional surveys at least quarterly in all individuals enrolled in the project. Blood was taken by finger prick and we examined 200 oil-immersion fields. We measured the parasite: leukocyte ratio for each plasmodial species and we enumerated separately the gametocytes of P. falciparum.
Between June 1990 and December 2010, four first-line drugs regimens were successively used for antimalarial treatment: oral quinine (Quinimax®) (October 1990–December 1994), chloroquine (January 1995–October 2003), sulfadoxine/pyrimethamine+amodiaquine (SP+AQ) (November 2003–May 2006) and artesunate+amodiaquine (AS+AQ) (June 2006–December 2010). Antimalarials were systematically given to young children in case of fever associated with a parasite: leukocyte ratio ≥2 [22] (link). When parasitemia was lower, the requirement for antimalarial treatment was decided taking into account all the patient's clinical, biological and epidemiological data [22] (link). Among older children (≥10 years) and adults permanently living in the village, it was rapidly observed that clinical malaria attacks lasted only a few hours even in cases where specific malaria treatment was delayed or not taken [23] (link), and thus in most cases (except pregnant women) only symptomatic treatment was given under close clinical surveillance (three daily visit at home until recovery) in order to reduce the selection of drug resistant malaria parasites. Urine tests carried out to detect the presence of antimalarials indicated that almost all positive results (>99%) were explained by treatments given in our clinic [22] (link). There were no chemoprohylaxis, intermittent preventive treatment nor presumptive malaria treatment in children or adults during the time period 1990–2010. At the beginning of the project, 48.6% (children: 51.1%, adults: 47.1%) of the villagers used traditional mosquito nets, which were untreated, and this proportion remained almost unchanged until July 2008 when LLINs were distributed to all villagers.
Full text: Click here
Publication 2012
Adult Amodiaquine Antimalarials Artesunate Biopharmaceuticals Bites Blood Body Temperature Child Chloroquine Culicidae Fever Fingers Households Leukocytes Malaria Marshes Mosquito Nets Oil Fields Parasitemia Parasites Pharmaceutical Preparations Plasmodium Pregnant Women Quinimax Quinine Submersion sulphadoxine-pyrimethamine Transmission, Communicable Disease Treatment Protocols Urinalysis Youth

Most recents protocols related to «Amodiaquine»

To investigate the potential of BI 425809 to reversibly inhibit the major human CYPs, CYP-selective substrates (phenacetin 60 μM [CYP1A2], bupropion 80 μM [CYP2B6], amodiaquine 2 μM [CYP2C8], diclofenac 5 μM [CYP2C9], S-mephenytoin 80 μM [CYP2C19], dextromethorphan 5 μM [CYP2D6], midazolam 2 μM, and testosterone 50 μM [CYP3A4/5]) were incubated with human liver microsomes and BI 425809 (0.015, 0.046, 0.137, 0.411, 1.23, 3.70, 11.1, 33.3, and 100 μM). For positive control reactions, BI 425809 was replaced with a CYP-selective inhibitor (α-naphthoflavone [CYP1A2], ticlopidine [CYP2B6], montelukast [CYP2C8], sulfaphenazole [CYP2C9], benzylnirvanol [CYP2C19], quinidine [CYP2D6], and itraconazole [CYP3A4/5]). Substrate metabolites were quantified with liquid chromatography–tandem mass spectrometry using gradient elution (mobile phase for amodiaquine metabolite—A, 5 mM ammonium formate in water/formic acid [100:0.1, v/v]; B, acetonitrile/formic acid [100:0.1, v/v]; mobile phase for all other substrate metabolites—A, water/formic acid [100:0.1, v/v]; B, acetonitrile/formic acid [100:0.1, v/v]) on a Synergi Hydro RP column (50 × 2.0 mm, 4 μm; Phenomenex) with positive electrospray ionization.
IC50 values were obtained using a 3-parameter dose-response, 4-parameter dose-response, or normalized dose-response model; model comparisons were performed in Prism 6 (GraphPad Inc) to determine the optimal model for each data set. A least-squares fitting approach was used, and the Hill slope was not constrained for the 4-parameter model.
Publication 2023
acetonitrile Amodiaquine BI 425809 Bupropion Cardiac Arrest CYP1A2 protein, human CYP2C8 protein, human CYP2C19 protein, human Cytochrome P-450 CYP2D6 Cytochrome P-450 CYP3A4 Dextromethorphan Diclofenac formic acid formic acid, ammonium salt Homo sapiens Itraconazole Liquid Chromatography Mephenytoin Microsomes, Liver Midazolam montelukast Phenacetin prisma Quinidine Sulfaphenazole Tandem Mass Spectrometry Testosterone Ticlopidine
The inhibition of the conversion of
a specific substrate to its metabolite was assessed at 37 °C
using human liver microsomes and to determine the inhibition of cytochrome
P450 isoenzymes by a test compound. For the following cytochrome P450
isoenzymes, turnover of the respective substrates was monitored: CYP3A4:
Midazolam; CYP2D6: Dextromethorphan; CYP2C8: Amodiaquine; CYP2C9:
Diclofenac; CYP2C19: Mephenytoin; CYP2B6: Bupropion; CYP1A2: Tacrine.
The final incubation volume contained TRIS buffer (0.1 M), MgCl2 (5 mM), human liver microsomes dependent on the P450 isoenzyme
measured (ranging from 0.05 to 0.5 mg/mL), and the individual substrate
for each isoenzyme (ranging from 1 to 80 μM). The effect of
the test compound on substrate turnover was determined at five concentrations
in duplicate (e.g., highest concentration 50 μM with subsequent
serial 1:4 dilutions) or without test compound (high control). Following
a short preincubation period, reactions were started with the co-factor
(NADPH, 1 mM) and stopped by cooling the incubation down to 8 °C,
followed by addition of one volume of acetonitrile. An internal standard
solution is added after quenching of incubations. Peak area of analyte
and internal standard is determined via LC-MS/MS. The resulting peak
area ratio of analyte to internal standard in these incubations is
compared to a control activity containing no test compound to determine
the inhibitory IC50.
Full text: Click here
Publication 2023
acetonitrile Amodiaquine Bupropion CYP1A2 protein, human CYP2C8 protein, human CYP2C19 protein, human Cytochrome P-450 CYP2D6 Cytochrome P-450 CYP3A4 Cytochrome P450 Cytochromes Dextromethorphan Diclofenac Homo sapiens Isoenzymes Magnesium Chloride Mephenytoin Microsomes, Liver Midazolam NADP Psychological Inhibition Tacrine Tandem Mass Spectrometry Technique, Dilution Tromethamine
As previously described, the CYP-mediated metabolic stability was evaluated as test compounds (with a final concentration of 1 μM) incubated with pooled rat or human liver microsomes (0.2 mg/mL protein) in 100 mM of potassium phosphate buffer with 3 mM of MgCl2, at pH 7.4 [41 (link)]. After pre-incubating for 5 min at 37 °C, the reaction was initiated with NADPH (at a final concentration of 1 mM). Negative control without NADPH and positive control with cocktail probe compounds (phenacetin, diclofenac, S-mephenytoin, bupropion, amodiaquine, dextromethorphan, and midazolam) were conducted simultaneously. The AO-mediated metabolic stability was evaluated as test compounds (with a final concentration of 1 μM) incubated with 37 °C pre-incubated pooled human hepatocyte cytosol (0.5 mg/mL protein) in 100mM of potassium phosphate buffer at pH 7.4, with or without AO inhibitor raloxifene (with a final concentration of 5 μM) [42 (link)]. Aliquots from the incubations were removed at different time points in the duration of 60 min and added into 5×volume prechilled internal standard-acetonitrile solution to stop the reactions. In order to prepare the supernatant for LC-MS/MS analysis, the supernatant was centrifuged at 15,000× g for 10 min and stored at −20 °C.
Full text: Click here
Publication 2023
acetonitrile Amodiaquine Buffers Bupropion Cytosol Dextromethorphan Diclofenac Homo sapiens Magnesium Chloride Mephenytoin Microsomes, Liver Midazolam NADP NOS2A protein, human Phenacetin potassium phosphate Proteins Raloxifene Tandem Mass Spectrometry
This was a longitudinal, single-arm, prospective study to evaluate P. falciparum tolerance to ART and its derivatives in children with uncomplicated malaria aged 6 months to 14 years in 3 health facilities in the Greater Accra region of Ghana. The study focused mainly on day 3 post artemether-lumefantrine (AL) treatment parasitaemia, 72-h ex vivo RSA after dihydroartemisinin (DHA) exposure, 72-h parasite clearance in vitro against a panel of 6 drugs (ART, AS, artemether [AM], DHA, amodiaquine [AQ], lumefantrine [LUM], and the following molecular markers of drug tolerance / resistance: Single Nucleotide Polymorphisms (SNPs), Multiple Nucleotide Polymorphisms (MNPs), Insertions & Deletions (INDEL) in Pfk13, Pfcoronin, P. falciparum multidrug resistance protein 1 (Pfmdr1), multidrug resistance protein 2 (Pfmdr2), dihydrofolate reductase (Pfdhfr), dihydropteroate synthetase (Pfdhps), signal peptide peptidase (Pfspp), and multidrug resistance-associated protein 2 (Pfmrp2) genes. It sought to set up correlates of ART tolerance.
Full text: Click here
Publication 2023
ABCB1 protein, human Amodiaquine Artemether artenimol Biological Markers Child derivatives Dihydropteroate Synthase Genes Genetic Polymorphism Immune Tolerance INDEL Mutation Lumefantrine Lumefantrine, Artemether Malaria Multidrug-Resistance Associated Protein 2 Nucleotides P-glycoprotein 2 Parasitemia Parasites Pharmaceutical Preparations signal peptide peptidase Single Nucleotide Polymorphism Tetrahydrofolate Dehydrogenase
Using HLM as the enzyme source, the CYP inhibition potential was employed to assess the inhibitory effect of the TbB ligands on the CYP1A2, 2B6, 2C8, 2C9, 2C19, and 2D6 3A4 isoforms. Phenacetin (CYP1A2, 10 mM), tolbutamide (CYP2C9, 20 mM), bupropion (CYP2B6, 10 mM), mephenytoin (CYP2C19, 100 mM), midazolam (CYP3A4, 5 mM), amodiaquine (CYP2C8, 0.02 mM), and dextromethorphan (CYP2D6, 5 mM) were used as probe substrates. The test substance included TbB ligand (final concentrations of 10 M for each) and HLMs (final concentration of 0.1 mg protein per mL), with or without 1.0 mM NADPH. The reaction was started by adding pre-incubated 25 μ L of NADPH into all wells and incubate at 37 °C for 10 min for 3A4 and 20 min for remaining all isoforms. The reaction was stopped by adding 100 μ L of stop solution to all wells. The samples were centrifuged at 4000 rpm for 10 min at 4 °C. Supernatant (100 μ L) was withdrawn, mixed with 200 μ L of water followed by LC-MS/MS analysis.
Full text: Click here
Publication 2023
Amodiaquine Bupropion CYP1A2 protein, human CYP2C19 protein, human Cytochrome P-450 CYP2C8 Cytochrome P-450 CYP2D6 Cytochrome P-450 CYP3A4 Dextromethorphan Enzymes Ligands Mephenytoin Midazolam NADP Phenacetin Protein Isoforms Proteins Psychological Inhibition Tandem Mass Spectrometry Tolbutamide

Top products related to «Amodiaquine»

Sourced in United States, Germany
Amodiaquine is a laboratory chemical used as a reference standard in analytical testing. It is a synthetic anti-malarial drug that can be utilized in the analysis and quality control of pharmaceutical products. The core function of Amodiaquine is to serve as a reference material for the identification and quantification of this compound in various samples.
Sourced in United States, Germany, United Kingdom, China, France, Macao, Sao Tome and Principe, Switzerland, Italy, Canada, Japan, Spain, Belgium, Israel, Brazil, India
Chloroquine is a laboratory chemical primarily used as a research tool in biochemical and cell biology applications. It is a white, crystalline solid that is soluble in water. Chloroquine is commonly used in experiments to study cellular processes, such as autophagy and endocytosis, by inhibiting the function of lysosomes. Its core function is to serve as a research reagent for scientific investigations, without making any claims about its intended use.
Sourced in United States, Germany, Sao Tome and Principe
Dextromethorphan is a laboratory chemical compound used as a research tool. It is a dissociative anesthetic and cough suppressant. Dextromethorphan is commonly used in scientific research, but its specific applications and intended uses should not be extrapolated or interpreted beyond its core function as a laboratory product.
Sourced in United States, France, Germany
Mefloquine is a synthetic compound used in the production of laboratory equipment. It is a key component in the manufacture of certain types of analytical instruments and devices used for research and testing purposes.
Sourced in United States, China, Germany, United Kingdom, Poland
Phenacetin is a chemical compound used in the manufacturing of various pharmaceutical and laboratory products. It serves as a key ingredient in the production process. Phenacetin has specific functional properties that make it a valuable component in relevant applications, but a detailed description of its core function is beyond the scope of this response.
Sourced in United States
Amodiaquine dihydrochloride dihydrate is a chemical compound used as a laboratory reagent. It is a crystalline solid that is soluble in water and other polar solvents. The compound has a molecular formula of C20H22Cl3N3O·2H2O.
Sourced in United States
N-desethylamodiaquine is a laboratory reference standard used for analytical testing and research purposes. It is a metabolite of the antimalarial drug amodiaquine. As a reference standard, it provides a well-characterized chemical compound for use in calibrating analytical instruments and verifying the identity and purity of samples.
Sourced in United States, Germany
Bupropion is a laboratory equipment product manufactured by Merck Group. It is a chemical compound used in various scientific and research applications. Bupropion is utilized for its specific chemical properties and functions, but a detailed description cannot be provided while maintaining an unbiased and factual approach.
Sourced in United States, Germany
Lumefantrine is a laboratory equipment product manufactured by Merck Group. It is a synthetic antimalarial drug used for the treatment of malaria.
Sourced in United States, United Kingdom, China, Belgium, Germany, Canada, Portugal, France, Australia, Spain, Switzerland, India, Ireland, New Zealand, Sweden, Italy, Japan, Mexico, Denmark
Acetonitrile is a highly polar, aprotic organic solvent commonly used in analytical and synthetic chemistry applications. It has a low boiling point and is miscible with water and many organic solvents. Acetonitrile is a versatile solvent that can be utilized in various laboratory procedures, such as HPLC, GC, and extraction processes.

More about "Amodiaquine"

Amodiaquine is a 4-aminoquinoline antimalarial medication used to treat and prevent malaria caused by the Plasmodium falciparum parasite.
It is effective against chloroquine-resistant strains and has a similar chemical structure to chloroquine, but a longer half-life and different metabolic profile.
Amodiaquine's mechanism of action involves interfering with heme detoxification within the parasite's digestive vacuole.
Despite its efficacy, the use of amodiaquine has been limited due to safety concerns, including the risk of liver toxicity and agranulocytosis with prolonged treatment.
Researchers must carefully optimize amodiaqine research protocols to ensure reproducibility and accuracy of results.
Amodiaquine is related to other antimalarial drugs like chloroquine, mefloquine, and lumefantrine.
It can also interact with medications like dextromethorphan, bupropion, and acetonitrile.
Understanding these relationships and potential interactions is crucial for safe and effective amodiaquine research and treatment.
PubCompare.ai's AI-driven platform can help streamline amodiaquine research by locating the best protocols from literature, preprints, and patents, and providing AI-driven comparisons to identify the most reliable and effective methods.
Leveraging this cutting-edge solution can help researchers optimize their amodiaquine studies for reproducibility and accuracy.