Science topic

Pharmacokinetics - Science topic

Pharmacokinetics are dynamic and kinetic mechanisms of exogenous chemical and drug ABSORPTION; BIOLOGICAL TRANSPORT; TISSUE DISTRIBUTION; BIOTRANSFORMATION; elimination; and TOXICOLOGY as a function of dosage, and rate of METABOLISM. It includes toxicokinetics, the pharmacokinetic mechanism of the toxic effects of a substance. ADME and ADMET are short-hand abbreviations for absorption, distribution, metabolism, elimination and toxicology.
Questions related to Pharmacokinetics
  • asked a question related to Pharmacokinetics
Question
3 answers
I did a pharmacokinetics for my compound and i used a sparse method in order to calculate PK parameters but i am not confident using NCA so my question is it possible to utilize sparse data in a compartmental analysis ?
Relevant answer
Answer
Jagtar Singh So i will apply population PK not non-population PK study, Is it right ?
  • asked a question related to Pharmacokinetics
Question
2 answers
I only have pharmacokinetic properties of each drug from drugbank
Relevant answer
Answer
Do you want to include in the model some interactions between the drugs. Are they impactinh each other's PK?
If not and you simply want to model the 3 drugs on its own it may be easier to model each seprately. You could also write a model with distinct parameters names for each drug, but this may become uneccesarily complictaed.
  • asked a question related to Pharmacokinetics
Question
7 answers
I can fathom the idea of PK being different b/w dogs and rodents, but why would I be seeing a significantly different PK profile for the same drug tested on rats and mice?
Relevant answer
Answer
Dear Colleague,
I hope this message finds you well. Understanding the pharmacokinetic (PK) differences between rats and mice is essential for interpreting preclinical data and translating findings to human applications. Several factors contribute to these differences, which I detail below:
  1. Metabolic Rate:Basal Metabolic Rate: Mice generally have a higher basal metabolic rate compared to rats. This can lead to faster drug metabolism and clearance in mice, impacting drug half-life and bioavailability.
  2. Enzyme Expression:Cytochrome P450 Isoenzymes: The expression levels and activity of cytochrome P450 enzymes can differ significantly between rats and mice. These enzymes play a crucial role in the metabolism of many drugs, influencing the rate of biotransformation and the formation of metabolites. Phase II Enzymes: Differences in the expression of phase II enzymes (e.g., glucuronidation and sulfation enzymes) also contribute to variations in drug conjugation and elimination.
  3. Absorption:Gastrointestinal Differences: Variations in gastrointestinal pH, transit time, and the expression of transporters and enzymes in the gut can affect the absorption rate and extent of orally administered drugs.
  4. Distribution:Body Composition: Differences in body fat composition and tissue distribution can influence the volume of distribution (Vd) of lipophilic drugs. Rats and mice may have different Vd values, affecting drug concentration in tissues. Plasma Protein Binding: Variations in plasma protein binding between species can alter the free (active) drug concentration, impacting the drug’s pharmacodynamics and kinetics.
  5. Excretion:Renal Function: Differences in renal blood flow, glomerular filtration rate (GFR), and tubular secretion between rats and mice can affect the excretion rate of drugs and their metabolites. Biliary Excretion: Species-specific differences in biliary excretion can influence the elimination of drugs that are primarily excreted via the bile.
  6. Physiological and Anatomical Differences:Organ Size and Function: Variations in the size and function of organs involved in drug metabolism and excretion (e.g., liver, kidneys) can impact pharmacokinetic profiles. Blood-Brain Barrier: Differences in the permeability of the blood-brain barrier can affect the distribution of drugs to the central nervous system.
  7. Genetic Factors:Strain-Specific Differences: Genetic variability between different strains of rats and mice can result in differences in drug metabolism and response. It is important to consider strain-specific characteristics when comparing PK data.
  8. Experimental Conditions:Housing and Diet: Variations in housing conditions, diet, and handling can influence physiological parameters and, consequently, drug pharmacokinetics. Standardizing these conditions is crucial for minimizing variability. Administration Route and Formulation: The route of administration (e.g., oral, intravenous) and the formulation of the drug (e.g., solution, suspension) can lead to differences in absorption and bioavailability between species.
In conclusion, multiple factors, including metabolic rate, enzyme expression, absorption, distribution, excretion, physiological and anatomical differences, genetic factors, and experimental conditions, contribute to the pharmacokinetic differences observed between rats and mice. Careful consideration and control of these factors are essential for accurate interpretation and comparison of PK data across species.
Should you have any further questions or require additional assistance, please feel free to reach out.
What factors may contribute to the PK differences in rats and mice?
  • asked a question related to Pharmacokinetics
Question
1 answer
Pharmacokinetic and pharmacodynamic differences, adjustments drug dosages in children to ensure safe and effective therapy.
Relevant answer
Answer
Medicine gets absorbed faster , spread diff due to more body water so we use weights, height, blood test , breakdown slower because of immature enzymes processes and paths under development
  • asked a question related to Pharmacokinetics
Question
2 answers
What are the pharmacokinetic properties of warfarin, and
how do they influence dosing and administration
considerations?
Relevant answer
Answer
  • asked a question related to Pharmacokinetics
Question
1 answer
Relevant answer
Answer
After oral administration, olanzapine is rapidly absorbed, reaching peak plasma concentrations within 5 to 8 hours. It has a high bioavailability of roughly 60%, with meals slightly slowing absorption. Olanzapine binds to plasma proteins and distributes throughout the body, including passing the blood-brain barrier to affect the central nervous system. Metabolism occurs largely in the liver via the cytochrome P450 system, specifically CYP1A2 and CYP2D6 enzymes, producing a variety of metabolites, including the active metabolite N-desmethylolanzapine. Then, elimination is predominantly accomplished through hepatic metabolism and subsequent renal excretion of metabolites, with a half-life of around 30 hours.Patients with hepatic or renal impairment may require dosage changes due to changed pharmacokinetics.
  • asked a question related to Pharmacokinetics
Question
1 answer
Describe the pharmacokinetic and pharmacodynamic changes that affect drug metabolism and response in pregnant patients. Pharmacokinetic and pharmacodynamic changes occur during pregnancy, impacting drug metabolism, distribution, and response in pregnant patients. Understanding these changes is crucial for safe and effective medication management during pregnancy.
Relevant answer
Answer
Pharmacokinetic and pharmacodynamic changes occur during pregnancy, impacting drug metabolism, distribution, and response in pregnant patients. Understanding these changes is crucial for safe and effective medication management during pregnancy. Here's an overview of the key alterations:
  1. Pharmacokinetic Changes:a. Absorption:Gastric emptying may be delayed due to hormonal changes and mechanical factors, leading to altered absorption rates for orally administered drugs. Increased gastric pH and decreased gastrointestinal motility can affect drug dissolution and absorption, particularly for weakly acidic drugs. b. Distribution:Increased maternal blood volume and cardiac output during pregnancy result in higher plasma volume and expanded extravascular fluid compartments. Protein binding may be altered due to increased levels of circulating binding proteins (e.g., albumin), affecting the free fraction of drugs. Lipid solubility and placental transfer characteristics influence the distribution of drugs across the placenta and into fetal circulation. c. Metabolism:Hepatic blood flow and enzyme activity may increase during pregnancy, affecting drug metabolism by cytochrome P450 enzymes. Induction or inhibition of specific metabolic pathways can alter the clearance of drugs, leading to changes in plasma concentrations and therapeutic effects. Phase II conjugation reactions (e.g., glucuronidation, sulfation) may be enhanced or impaired, affecting the elimination of drugs and their metabolites. d. Elimination:Renal blood flow and glomerular filtration rate (GFR) increase during pregnancy, resulting in enhanced renal clearance of drugs. Changes in renal tubular function and urinary pH can impact drug excretion rates and half-life. Hepatic clearance may be altered due to changes in enzyme activity, protein binding, and biliary excretion pathways.
  2. Pharmacodynamic Changes:a. Sensitivity to Drugs:Hormonal fluctuations and physiological changes during pregnancy can influence drug sensitivity and response. Changes in receptor density, affinity, or downstream signaling pathways may affect drug efficacy and potency. b. Fetal Effects:Drugs that cross the placenta can have direct effects on fetal development and physiology. Teratogenicity, fetal growth restriction, and developmental abnormalities may occur if drugs interfere with critical pathways or organogenesis. c. Maternal Response:Pregnancy-induced alterations in cardiovascular, respiratory, and renal function can impact the maternal response to drugs. Hemodynamic changes may affect drug distribution, metabolism, and elimination, leading to variations in pharmacokinetic parameters and clinical outcomes.
In summary, pharmacokinetic and pharmacodynamic changes during pregnancy can influence drug metabolism, distribution, and response, posing challenges for medication management in pregnant patients. Clinicians must consider these alterations when prescribing medications, adjusting dosages, and monitoring therapeutic effects to optimize maternal and fetal outcomes while minimizing risks of adverse drug reactions. Close collaboration between obstetricians, anaesthesiologists, and pharmacists is essential for safe and effective medication use during pregnancy.
  • asked a question related to Pharmacokinetics
Question
1 answer
Explain the physiological changes that occur during pregnancy and their implications for anaesthesia management. During pregnancy, significant physiological changes occur to support fetal development and accommodate the needs of both the mother and the growing fetus. These changes have implications for anaesthesia management and can impact various organ systems and pharmacokinetics.
Relevant answer
Answer
During pregnancy, significant physiological changes occur to support fetal development and accommodate the needs of both the mother and the growing fetus. These changes have implications for anaesthesia management and can impact various organ systems and pharmacokinetics. Here's an explanation of some key physiological changes and their implications for anaesthesia:
  1. Cardiovascular System:Increased Blood Volume: Blood volume increases by approximately 30-50% by the end of pregnancy to meet the demands of the developing fetus and placenta. Increased Cardiac Output: Cardiac output increases due to the rise in blood volume, heart rate, and stroke volume, reaching its peak in the third trimester. Implications for Anaesthesia: Pregnant patients are at increased risk of hypotension and hemodynamic instability during anaesthesia due to the increased blood volume and cardiac output. Close monitoring of blood pressure, fluid status, and cardiac function is essential to prevent maternal and fetal compromise.
  2. Respiratory System:Increased Oxygen Consumption: Oxygen consumption increases by approximately 20% during pregnancy to meet the metabolic needs of the mother and fetus. Decreased Functional Residual Capacity (FRC): FRC decreases due to upward displacement of the diaphragm by the growing uterus, leading to decreased lung volumes and increased risk of atelectasis. Implications for Anaesthesia: Pregnant patients have reduced respiratory reserve and are more susceptible to respiratory compromise, especially during induction of anaesthesia and airway manipulation. Adequate preoxygenation and rapid sequence induction techniques may be necessary to minimize the risk of hypoxemia.
  3. Renal System:Increased Renal Blood Flow and Glomerular Filtration Rate (GFR): Renal blood flow and GFR increase to accommodate the increased metabolic demands and eliminate metabolic waste products. Decreased Ureteral Tone: Ureteral relaxation and compression by the gravid uterus increase the risk of urinary stasis and hydronephrosis. Implications for Anaesthesia: Pregnant patients are more prone to fluid retention, electrolyte imbalances, and renal dysfunction. Careful fluid management and monitoring of renal function are important to prevent volume overload and electrolyte disturbances.
  4. Gastrointestinal System:Delayed Gastric Emptying: Delayed gastric emptying and decreased lower esophageal sphincter tone increase the risk of aspiration during pregnancy. Increased Risk of Gastroesophageal Reflux: Hormonal changes and mechanical factors contribute to gastroesophageal reflux and heartburn in pregnant patients. Implications for Anaesthesia: Pregnant patients are at increased risk of aspiration pneumonitis during induction of anaesthesia and airway manipulation. Preoperative fasting and administration of gastric acid-reducing medications (e.g., H2 blockers, proton pump inhibitors) may be necessary to reduce the risk of aspiration.
  5. Hormonal Changes:Increased Levels of Progesterone and Relaxin: Progesterone and relaxin levels rise during pregnancy, leading to relaxation of smooth muscle tone, ligament laxity, and increased joint mobility. Implications for Anaesthesia: Pregnant patients may have increased susceptibility to nerve injury, musculoskeletal strain, and joint dislocation during positioning for surgery and neuraxial anaesthesia. Careful positioning and padding are essential to minimize the risk of maternal and fetal injury.
In summary, the physiological changes that occur during pregnancy have significant implications for anaesthesia management, affecting cardiovascular, respiratory, renal, gastrointestinal, and hormonal systems. An understanding of these changes is essential for anaesthesia providers to optimize maternal and fetal outcomes and minimize perioperative complications in pregnant patients undergoing surgery or obstetric procedures. Close monitoring, individualized care, and multidisciplinary collaboration are key components of safe and effective anaesthesia management during pregnancy.
  • asked a question related to Pharmacokinetics
Question
3 answers
I want to do mathematical modelling of Pharmacokinetic data from plasma drug concentration in mice. Can someone please suggest me any freely available software for PK Modelling.
Relevant answer
Answer
Hi,
If you are a University scientist try to ask Lixoft about PKanalix availability (academic license) ..... easy intuitive soft (https://lixoft.com/).
I also recommend their spring school (every year PK training program online, free of charge).
Best regards,
Tomasz
  • asked a question related to Pharmacokinetics
Question
1 answer
I am new to the field and I am currently working with lenvatinib. I have found that different research groups used different vehicles for lenvatinib in mouse experiments. I have even found out that some research groups did not mention the vehicle they used. I am so confused. Which vehicle should I use? For example, one group of researchers used water to prepare 3 mg/mL lenvatinib solution. (Mizuo, H., Mano, Y. Cross-species comparison in nonclinical pharmacokinetics of lenvatinib by a simple HPLC with ultraviolet detection. Sci Rep 13, 8349 (2023). https://doi.org/10.1038/s41598-023-35297-z). I tried it, but the powder did not dissolve. Can anyone help?
Relevant answer
Answer
Carboxymethylcellulose (CMC)
  • asked a question related to Pharmacokinetics
Question
1 answer
Say you have been asked to design an experiment to complete a pharmacokinetic profile of a new antidepressant drug, How would you go about deciding what doses to use for (Oral / IV) administration in an in-vivo animal model using rats for example? Providing you have no toxicology information, would you use dosages that have been used in other anti-depressant studies? or is there a general rule for minimum / maximum doses being used for PK studies?
Relevant answer
Answer
Dear friend Jake Ellis-Williams
Alright, let's tackle this like I would!
Designing a pharmacokinetic study for a new CNS drug, eh? Now, we're talking real science. First things first, if you Jake Ellis-Williams don't have any toxicology data, you're in a bit of a tight spot. But I dont shy away from challenges!
1. **Start Low, Go Slow:**
- In the absence of toxicology data, the golden rule is to start low. You Jake Ellis-Williams don't want to accidentally turn your rats into rocket ships, do you? Use a dose that's unlikely to cause adverse effects.
2. **Reference Similar Compounds:**
- Check out similar drugs in the antidepressant family. It's not a perfect science, but it gives you Jake Ellis-Williams a ballpark figure. Look at the literature, see what doses they used, and take a leap from there.
3. **Consider Route of Administration:**
- Are you Jake Ellis-Williams going oral or intravenous? Oral is more common, but if you're feeling adventurous, go IV. Adjust your dose accordingly; IV doses are typically lower due to higher bioavailability.
4. **Species Matters:**
- Rats are not humans. Shocking, I know. Their metabolism differs, so don't just scale up a human dose. Consider allometric scaling, a fancy term for adjusting doses based on body surface area.
5. **Monitor Behavior:**
- Keep a close eye on your rats. If they start tap-dancing or speaking French, you Jake Ellis-Williams might have gone a bit too high. Watch for signs of toxicity and adjust your doses accordingly.
6. **Pilot Studies are Your Friends:**
- Before diving into the big show, run some pilot studies. Test the waters with a few rats. It's like a movie trailer – gives you Jake Ellis-Williams a taste without committing to the full feature.
7. **Consult the Oracle (Experienced Scientists):**
- If you Jake Ellis-Williams have access to seasoned scientists, consult them. They've been around the block and can provide invaluable insights. Wisdom often comes with gray hair and lab coats.
8. **Ethics Committee Approval:**
- Don't forget to run your doses past the ethics committee. They might not have a Ph.D., but they know a thing or two about keeping experiments humane.
Remember, my science-seeking friend, this isn't a one-size-fits-all scenario. The key is to be cautious, use your scientific instincts, and be ready to adapt as your data rolls in. May the pharmacokinetics odds be ever in your favor!
  • asked a question related to Pharmacokinetics
Question
2 answers
In the paper "Pharmacokinetics and metabolism of N-[N-[3-(3-hydroxy-4-methoxyphenyl) propyl]-α-aspartyl]-L-phenylalanine 1-methyl ester..." (Food Chem Toxicol. 2011 Nov;49 Suppl 1:S8-29) is reported that advantame is metabolized to ANS9801-acid.
However, the structure for ANS9801-acid looks the same as that I have found for advantame in spiderchem, as shown in the picture.
What is the structure of ANS9801-acid, also known as de-esterified advantame?
Relevant answer
Answer
The abstract of the paper you mention states:
Data indicated that absorption was mainly as ANS9801-acid (de-esterified advantame), which was formed in the gastrointestinal tract as a result of the hydrolysis of the methyl ester group of the parent compound.
So, in the picture, you provided ANS9801 is equivalent to Advantame (if we omit the water molecule included in Advantame). But the paper clearly mentions ANS9801-acid, that indicates the structure of ANS9801 without methyl ester functionality (which has been hydrolyzed to free acid, meaning that ANS9801-acid has, in fact, two carboxylic groups).
The exact structure is provided and subscribed as ANS9801-acid in Fig.22, showing the proposed metabolic pathway of ANS9801, in the paper you cite.
  • asked a question related to Pharmacokinetics
Question
4 answers
I have a few samples and I want to evaluate pharmacokinetics and drug-likeness for the publication, which web server or tool is accepted by the publisher.
Relevant answer
Answer
pkcsm and AdmetSAR are more often applied webservers for evaluating ADME properties, Protox II for toxicity, and swissADME for drug-likeness properties. If you have access to Schrodinger software, it will be better to use Quikprop so as to evaluate the pharmacokinetic properties comprehensively.
  • asked a question related to Pharmacokinetics
Question
3 answers
Plasma drug concentration and brain distribution study
Relevant answer
Answer
Try discus the topic with Lixoft they have amazing software and free of charge academic version probably is a possible option. Link below:
  • asked a question related to Pharmacokinetics
Question
7 answers
I am planning to conduct pharmacokinetic studies in mice. Considering the availability of low blood volumes per mice, how to collect the samples efficiently in time frames of 5 min, 15 min, 30 min, 1 h, 2h, 4h, 6h, 8h, and 12h? The total blood volume in a mice weighing 25-28 gm is hardly 2 ml.
Relevant answer
Answer
Hi Jagtar, it would depend on what is the lower limit of quantification of your analytical method and subsequently how much blood you need per sampling time point. Generally for a 25 g mouse the recommendation is not more than 250uL blood in 24h period. I would recommend that if you need 250uL per time point (around 5 drops) then you could make groups of 3-5 mice for each time point. ie. dose 18-45 mice. Alternatively to reduce the number of mice in the study, you could refer literature available for the drug and reduce your timepoints.
  • asked a question related to Pharmacokinetics
Question
3 answers
my work on molecular docking, pharmacokinetics, DFT
KINDLY SUGGEST
Relevant answer
Answer
BUT THIS JOURNAL IS TAKING CHARGE/FEE FOR PUBLISHING ARTICLE.
PLEASE SUGGEST REPUTED JOURNAL WHICH WILL NOT TAKE CHARGE.
  • asked a question related to Pharmacokinetics
Question
4 answers
Does somebody has a pharmacokinetics model available in Excel? I’m working with a lot medications and would very much like to know how it builds up and to what extent. A simple model suffices but assume that multiple administration is required. Thanks very much, Marcel
Relevant answer
Answer
yes i worked two times but this model not properly works in excel.This model given my senior.After i used other software.
  • asked a question related to Pharmacokinetics
Question
4 answers
Dear Kinetic experts,
I need one kind suggestion from you. One X compound with molecular weight of 200.1, is highly water soluble with oral bio-availability of 65-75%. In that case I want to achieve 1 micro mole in plasma (irrespective to protein bound %) for that how much dose I should give by orally. Is that any standard formula is available for this calculation?
Please note that in my study design, if the compound reaches more than 1.5 micro mole it will become inactive. So specific concentration in plasma is really challenge. Please help with this
Thanks
Relevant answer
Answer
Pl refer link (page 22-23) for calculation of loading and maintenance dose based on PK parameters.
Regards
Hitesh Chavda
  • asked a question related to Pharmacokinetics
Question
13 answers
In the article titled "FDOPA-(18F): a PET radiopharmaceutical recently registered for diagnostic use in countries of the European Union", two half-lives are reported for FDOPA.
"6-fluoro-(18F)-L-dopa is removed according to a bi-exponential kinetic process with biological half-lives of 12 hours (67-94 %) and 1.7 - 3.9 hours (6-33 %). Both these half-lives appear to be age-dependent. The 18F-activity is excreted through the kidneys, 50 % with a half-life of 0.7 hours and 50 % with a half-life of 12 hours.
On basis of these data, a biokinetic model for 6-fluoro-(18F)-L-dopa was developed. This model assumes that 100 % of the 18F activity is homogeneously distributed in the body and eliminated through the kidneys with biological half-lives of 1 hour (50 %) and 12 hours (50 %). This model was considered to be independent of age."
What does this mean?
Relevant answer
Answer
David Salehi Thank you for your recommendations. I will certainly have a look at them.
  • asked a question related to Pharmacokinetics
Question
5 answers
Someone was taking Daflon 500 mg (450 mg Diosmin, 50 mg Hesperidin) twice daily for her varicose veins. Now, the manufacturer makes Daflon 1000 mg (900 mg Diosmin, 100 mg Hesperidin).
Now, the Q is, if she takes the new dosage form 1000 mg once daily, will this dose be equivalent to 500 mg once daily or the pharmacokinetics and the therapeutic effects will be different?
Relevant answer
Answer
based on a double-blind, multicenter, RCT comparing 1,000 mg tabs vs 500 mg tabs (same daily dose) in acute hemorrhoid, the authors concluded that "We have been demonstrated that the new single MPFF 1000 mg tablet has clinical acceptability and a good safety profile, comparable to that of MPFF 500 mg tablets. MPFF 1000 mg was as effective as MPFF 500 mg in reducing anal pain and bleeding. The new dose regimen should lead to better treatment adherence and consequently to better management of hemorrhoidal disease."
  • asked a question related to Pharmacokinetics
Question
3 answers
I have some synthesized drug and want to study the pharmacokinetic of this drug. But I am confused with the dosing calculation, how much dose i should administer to animal especially rat for the evaluation of pharmacokinetic evaluatioion.
Relevant answer
Answer
Hi,
Sounds like a basic question but is very often asked by experienced scientists. The answer is it depends on your goal. Measurement PK does not always mean analysis 'only' PK so the dose will be different if:
  • specific aspects of the PK profile are analyzed (or model establishment)
  • pharmacokinetics or toxicokinetics
  • PK variability
  • comparison with homolog or another route of adm
  • route of administration (bioavailability)
  • presence of specific effects like flip-flop kinetics
  • double peak phenomena after a single dose
  • local/general tolerance
  • pilot analysis
  • number of doses
  • range of linear PK
  • cumulation, deep compartments
  • first, the pass effect
  • percent of blood plasma proteins binding
  • study item (biologics, biotech, prodrugs, synthetics)
  • additional PD observations
  • immunogenicity if appropriate (ADA, NAbs control)
And many others factors and goals may change decisions about dose amount. But one critical is a range of validation of your analytical method. If for example, LLOQ of your method is 10 ng/mL you can't use a dose that gives concentrations between 0.01-3.0 ng/mL. Consequently in your PK study:
  • Cmax should be slightly lower (variability of PK) than the HLOQ of the method
  • Clast should be slightly higher than the LLOQ of the method
  • Clast should be close or more than 20 x lower than Cmax
  • If blood volume is 7% of the BW then you can calculate concentration at time point = 0 (Cmax if the iv bolus is used) based on this volume and study item amount you can calculate more less concentration close to Cmax. In the rat (300 g) blood volume is circa 21 mL.
Please remember that the calculation is very very general because depending on study item many processes may change this value within seconds (blood protein binding, first-pass effect, route of administration, and presence of absorption phase etc). So if possible always if you have no data which can be translated from other animals or homologs try to make a pilot study it is always very helpful for both optimizations of the analytical method and dose level. The pilot could be done only with a few animals and a few samples of blood.
Some useful tips related to single-dose PK you can find in my paper below:
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
3 answers
Most free webservers that I am using for predicting the pharmacokinetics and biological activities of molecules are restricted to organic molecules only not exceeding over 1,200 in molecular weights. However, there are interesting biomolecules in nature that deserve attention for in silico studies which cannot be tested using these online tools.
Relevant answer
Answer
Hi, how about this site?
Compartment modeling: http://ca.kscpt.org/
  • asked a question related to Pharmacokinetics
Question
2 answers
For example
Times 0.5, 1, 2, 4, 6 hours and concentrations <2.50, <2.50, 7.80, 53.3, 279
I am using PKsolver the excel add in but I am not getting the expected value.
Relevant answer
Answer
I guess when concentration becomes too low, you might have to look for cues like calibration of the detector, and other variables such as temperature and pressure which affect rate of the reaction. Solubility and saturation might also be considerations. A slow reaction might take a longer period of time to measure.
  • asked a question related to Pharmacokinetics
Question
8 answers
I have 4 groups, each group is composed of 5 mice, I set up to withdraw at 7 intervals to complete my study. It is not possible to withdraw 7 samples from a single mouse so Is it scientifically right to withdraw 3-4 samples from all group members (5 mice) on a triplicate base and then calculate PK parameters, average them and calculate SD? Is that acceptable scientifically?
Relevant answer
Answer
Hi,
The best way for sampling from small animals is the sparse sampling method (if you have no access to cannulated animals + a very sensitive method). In the case of sparse sampling, you can implement the options present in software for example Phoenix WinNonlin (see reference below). Even if you work with very small animals or sparse sampling rules related to PK analysis are in close link with study design and PK curve structure. Those rules are the same like in humans or large animals If its typical single dose study with small population (excluding for example large populations and population PK were sampling is different or therapeutic drug monitoring etc.). Consequently, for example, 7 sampling points per typical single dose, frequent sampling curve in an exploratory study is not folowing the rules. Descriptiion of expectations related to typical frequent sampling in single dose PK studies (not population methods of analysis) you can find in my paper which shows some compilation of current consensus both in science and regulatory trials.
  • Minimum sampling points per PK curve: 12>18
  • Sampling points number from t0–tmax: >2-5 (if other than IV)
  • Time of sampling from t0–tlast: tmax + 3 x t1/2kel
  • Number of C–T for kel analysis: > 3-4
  • AUC% analysis (residual area): <20%
  • Clast: should be at level 100 x lower than expected mean Cmax
These are the principles that follow the principles of analytical geometry & mathematics. It is the common denominator of these rules. If you follow these rules, you will exclude any BIAS resulting from the lack of an optimized sampling design. The last rule about Clast Cmax will prevent you from calculating an elimination rate constant based on distribution phase rather than elimination. For example, if your expected Cmax is 100 ng/mL your analytical method LLOQ should be around 1 ng/mL rather than 10-15 ng/mL. If you follow these rules, you will always be on the safe side. To implement them, the best solution is a pilot analysis with a few animals before starting the main study and a validated sensitive method of analysis.
Hope this comment could be useful,
Best regards
Tomasz
.
  • asked a question related to Pharmacokinetics
Question
3 answers
I'm working on my master degree i'm measuring peak plasma concentrations for a drug (cmax) and will correlate it with a genetic marker but i made a mistake of not fixing a time interval for blood samples withdrawals i drawed the samples within a time range
Is there a way that i can use to correct real sampling times so i can use it and correlate it with the genetic marker
Relevant answer
Answer
Dear Sherouk,
Another thought I had is that you may be able to study the correlation differently.
Is the biomarker expected to change very rapidly in response to drug? Is this why you want to correlate to Cmax? Is it also going to be itself eliminated very quickly when drug concentrations decrease?
If these conditions are met you could try to fit an Emax / Imax model to the data (biomarker as a function of drug concentration at any time).
For this to makes sense you would need the biomarker to have a fast turnaround (faster than drug kinetics) and to show fast response to drug.
  • asked a question related to Pharmacokinetics
Question
5 answers
''Using the pharmacokinetic parameters and a pharmacokinetic-pharmacodynamic model, it is possible to predict the pharmacodynamic response to certain drugs; this provides useful information for understanding drug action and determining dosage regimen''. (Baggot, 1990)
Reference:
Baggot JD. Pharmacokinetic-pharmacodynamic relationship. Ann Rech Vet. 1990;21 Suppl 1:29S-40S.
Relevant answer
Answer
Respected Prof Faraz Khurshid , I cannot agree with you more. I think PD/PK are really subjects that needs to be emphasized in modern pharmacy training. We really need fundamental and basic scientific training for every pharmacist. Scientific concept is important for clinicians.
I also like evidence-based medicine, I guess this is really an important direction for our future generations. It is the black and white evidence that really underlie our decision and we should record the evidence accordingly so that we are speaking the same language and sharing our knowledge.
It is important that every decision is recorded and traceable, so that we are recognized as a professional and/or expert in a particular area(s).
  • asked a question related to Pharmacokinetics
Question
5 answers
Simple pharmacokinetic parameters of five potent compounds after a single oral administration to female mice.
how to review this pharmacokinetic results
Relevant answer
Answer
Hi Manikandan. Interesting exercise. Kindly see the attached workout. I hope it will guide your further efforts on the assignment.
In summary, it appears from the first table that:
1. Compounds 2 and 3 exhibit lower clearance, and higher bioavailability.
2. Compared to Compounds 1 and 5 (which exhibit equipotency), Compounds 2, 3, and 4 exhibit lower potency, Compound 2 much less so than the others.
3. Compound 4 exhibits lower bioavailability, a lower Cmax and, higher clearance.
4. Compound 5 exhibits the lowest Cmax but with unknown bioavailability and clearance. Safe to assume a high Vd and thus high clearance from the central compartment, and low bioavailability.
  • asked a question related to Pharmacokinetics
Question
1 answer
Is it possible to determine the correction factor (Km) to estimate the (AED) for Lepidopteran species? Haven't found any literature that discusses the MRSD for Lepidopteran species. Would very much appreciate it if someone has any insight into it. Need to calculate the drug dose for Bombyx Mori.
Relevant answer
Answer
Dear Siam,
I'm afraid I haven't come across allometry for such cases. In general allometry is used to scale between mammalian species (or individual of different sizes for pediatric applications).
The general idea is that flows (clearance) scales with a factor of about 0.75, so this applies also to dose (which is expected to be a function of clearance). So it follows a relationship of a*BW^0.75.
In absolute terms you could apply the formula with the weight of any species / individual. However, I suspect that the empirical principles mainly established between mammals may not apply to invertebrates.
To note that even between more similar species like mammals allometry does not always work well.
Out of curiosity, how do you apply drugs in insects? Can you actually apply orally or how it it done?
  • asked a question related to Pharmacokinetics
Question
3 answers
Did you ever experience a conceptual change in pharmacology where was a discrimination b/w phenomena/concepts that you previously regarded as a single phenomena/concept?
For instance, proton pump inhibitors are often thought to be interchangeable, but some differences have emerged in their pharmacological properties, which may be reflected in some aspects of clinical efficacy. Such differences include potency, speed of onset and duration of pH 'holding times' (2004)
Reference
Robinson M. Review article: the pharmacodynamics and pharmacokinetics of proton pump inhibitors--overview and clinical implications. Aliment Pharmacol Ther. 2004;20 Suppl 6:1-10. doi:10.1111/j.1365-2036.2004.02160.x
Relevant answer
Answer
The words selectivity, specificity, and sensitivity (derived from Latin seligere, specificus, sensitivus), can be confusing terms as they are often used synonymously in the medical literature. However, they should not be used
interchangeably as each represents a different phenomenon.
Reference:
Mencher, S. K., & Wang, L. G. (2005). Promiscuous drugs compared to selective drugs (promiscuity can be a virtue). BMC clinical pharmacology, 5(1), 1-7.
  • asked a question related to Pharmacokinetics
Question
3 answers
needing help in the criteria of choosing the drug to use when studying pharmacokinetics changes in patients with congestive heart failure
Relevant answer
Answer
many thanks doctors for your response..
would you recommend any supporting articles for this area of study Falguni Manoj Saraswat Tohlang Sehloho
  • asked a question related to Pharmacokinetics
Question
1 answer
As we know, polymyxin B contains many components (e.g. polymyxin B1, B2, etc.), do we have any evidence to show that those major components have different antimicrobial activity, pharmacokinetic property and propensity for toxicity?
Relevant answer
Answer
This paper could address the issue:
  • asked a question related to Pharmacokinetics
Question
2 answers
Simcor contains simvastatin and niacin. Simvastatin is metabolized by CYP3A4, and niacin is able to inhibit this isoform. Is niacin a strong or weak CYP3A4 inhibitor? Can niacin alter simvastatin pharmacokinetics?
Relevant answer
Answer
There are many iso enzymes that metabolism drug ,not necessarily the both Niacin and statin have same iso enzyme.
Important other important factor is the dose of Niacin may be very low to elicit metaboliic process.
  • asked a question related to Pharmacokinetics
Question
3 answers
In the pharmacokinetics and pharmacodynamics of frusemide
Relevant answer
Answer
@karel
I wish to know if there's any genetic polymorphism/variation in the pharmacology of frusemide. Whether at the level of safety, pk or pd?
Is there anything of concern in this regard sir?
  • asked a question related to Pharmacokinetics
Question
4 answers
I have data for concentration vs time for plasma and other tissues of the body for orally administered drug. I got analysis for plasma only using PK solver (Microsoft excel based Add-in) but not for other tissues.
Relevant answer
Answer
Hi Ashish,
You can do NCA analysis of all derived data if you have a full profile in organs.
Additionally from tissue data, you can derive tissue to plasma ratio. It will help you in understanding the drug disposition in various tissue.
From the CT profile of the animal, you can extrapolate the various doses and dose regimens by compartmental analysis. if you have sufficient subject then you can derive information about covariate impact on PK by popPK.
if you need further assistance let me know.
  • asked a question related to Pharmacokinetics
Question
8 answers
Hello,
I am having trouble interpreting some pharmacokinetic data. I work with mice. I have two groups of animals all given with the same drug in the same route at the same dosage. There were statistically significant differences in the AUC, Clearance, and Cmax between groups. However, the half-life was the same for all groups. Two of these groups were infected with a virus that replicates in the kidneys, so we expected them to have impaired renal clearance of the drug. If that were the case, wouldn't we see a prolonged t1/2? I am concerned that even though there are differences in the AUC and Clearance, without changes in T1/2, we can't conclude that the variation in PK parameters is due to changes in renal function.
Relevant answer
Answer
That makes sense. Could you be referring to C0 instead of Cmax instead? C0 is the extrapolated value to time 0, which will be Dose/Vd. If C0 changed and your dosing is constant, it is likely that there was a difference in volume of distribution that occurred. I generally find Vd harder to explain biologically as the reason for the change could be anything from changed transporters in blood cells to fat or other peripheral tissues or plasma protein binding.
In IV data, Ke = CL/Vd. If CL changed, but Ke did not, it is possible that Vd also changed to similar extent. Ke = ln2/(t1/2), so if Ke does not change, t1/2 will not change. An easy way to see this would be (as a quick and dirty way of ensuring your calculations were correct), if you plot your plasma data on a log 10 y axis, do the gradients of each mouse group look similar? Ke is pretty much the slope on that graph. Here is a list of useful PK equations that I often reference to understand such plots better. https://pharmacy.ufl.edu/files/2013/01/5127-28-equations.pdf
It is also possible that the virus you were administering did not do what you expected. Perhaps other things to investigate post sac, e.g. kidney histology - did the virus damage the kidney tissue?
  • asked a question related to Pharmacokinetics
Question
3 answers
Is there any software available to simulate the behaviour (growth, differentiation etc.) of different cell type on an implant surface.
Also, software related to simulation of drug loading and release from a drug carrier and its pharmacokinetics.
Relevant answer
Answer
Thank You Riaz & Tejas
  • asked a question related to Pharmacokinetics
Question
3 answers
I have harvested urine and serum at various time intervals from rats that have been given a crude extract . I want to assess the pharmacokinetics and pharmacodynamics of the individual compounds in the extract. However, I am not sure which method will work better for the pharmacokinetics because I do not know the proportion of each of the compounds in the extract.
Relevant answer
Answer
Hi,
Unfortunately, there are no such methods. Current solutions and models do not allow for selective analysis of one or several substances after administering the extract (.... which may contain several substances). Such analysis will only inform about interactions between constituents nothing else. Of course, the extent of these PK interactions will depend on the composition of the extract. What you can do is determine the concentration of the dominant substance in the extract and try to determine its concentration in blood samples or urine. However, the cognitive and scientific value of such an analysis will still be low. If an extract is administered, it is not possible to associate a single substance with a pharmacodynamic effect (PD). Currently, we do not have such data analysis capabilities. The effect of PD after administration of the extract is always the result of interactions between all components of the extract. We currently have neither software nor models that can separate these interactions from each other. Therefore, PK and PD are tested with the use of single selected substances or in systems where interactions with one substance are tested, but not with so many.
All you can do is try to characterize the chemical composition of the extract and describe/analyze the PD effect. Selective analysis of PK or selective PD analysis in your case is not possible.
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
7 answers
I am trying to evaluate PK profile (Plasma vs. time) of Test compounds in ICR mouse after PO administration as a screening study.
The common PK profile of these drugs indicates IV pattern, despite oral administration.
The meaning of the IV pattern that i mentioned is that the concentration of durgs in the plasma continuously decreases from first sampling time point (5 min) to the last sampling time point (24h).
In general, When administered with PO, it takes time for the drug to be absorbed, indicating a pattern in which the drug slowly increases and decreases.
When the drug is absorbed quickly after PO administration,
Can it indicate IV pattern?
Please give me advice
Relevant answer
Answer
Hi
There could be many reasons why you do not see typical absorption phase after oral administration in your PK profile for example:
  • Your test substance can be absorbed very quickly as there is a very efficient active transport system
  • The absorption phase is invisible because you are taking your first plasma samples too late after drug dosing
  • The drug may be administered by mistake with a tracheal probe, not in the oral cavity
  • Sometimes it is a problem with the carry-over in techniques such as HPLC UHPLC etc.
  • If mice are cannulated for blood sampling it may be the result of a poorly flushed cannula that was previously used for test substance administration
  • You definitely need to check whether there is a problem with the marking of samples.
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
5 answers
I am injecting intrathecally (between L5-L6) , in rats, cannabinoid analgesics and I was wondering if they can reach the brain and have effects there as well ?
Also I need to pretreat with antagonists (AM630 and Capsazepine) and was wondering how long does it take to get them cleared from spinal cord. I usually pretreat for 30min before agonist injections, is this too long or is it ok ? (we are interested in spinal effects only). I cannot inject the agonist and the antagonist simultaneously as the volume of each injection is too big to inject them one after the other (I had to dilute drugs in bigger volume in order to lower % of ethanol of solution: 20uL for antagonist and 30uL for agonist)
Relevant answer
Answer
Arthur Saus I agree with you
  • asked a question related to Pharmacokinetics
Question
4 answers
Nonlinear mixed-effects models (may) consider data below the limit of quantification (BLQ) in parameter estimation. However, an evaluation of the goodness-of-fit plots (observations vs predictions in particular, using spline interpolation), displays a strong trend (of spline interpolation, but not of the data) in the region of censored data, as if the model disregarded BLQ data and the data were the lower limit of quantification itself, as structured in the database. I believe that the database is structured correctly and that the model considered the censored interval. Apparently this plot is the only one to exhibit this behavior.
Is spline interpolation adequately representing the competence/capability of the final model in this case? How to handle this situation?
Relevant answer
Answer
Interpolation can be thought of in two ways:
1) Interpolation methods approximate some underlying model
2) Interpolation simply approximates a set of numbers continuously
In case 1), interpolating will capture the essence of the underlying function, if and only if, the data is representative of the fundamental model. Noise will definitely throw your results off.
In case 2), you are simply generating an approximation from a set of known numbers, e.g. Lagrange interpolation; forward, centered and backward Newtonian interpolation, &c. The numbers in hand may or may not represent anything in particular, or, they may not be accurate enough to enable one to gleam the underlying processes. Purely interpolated results can and do grow wild as the power of interpolation is increased.
What I suggest is a low power interpolation of the database, then look at the curve and compare to any model you have in mind. If the interpolated results resemble strongly enough some model, then you may further pursue investigating along that vein.
  • asked a question related to Pharmacokinetics
Question
5 answers
Friends,
I want more information about role of chiral drugs on drug delivery based on pharmacology, pharmacokinetics, pharmacodynamics, recepter binding, dose, potency , toxicity, safety with lot of examples. If you have any reference materials like article, book, or other formats and you please send to me.
Thanks you.
Relevant answer
Answer
Hi
"enantioselective pharmacokinetics" please check the keyword in google or NCBI.
geometrical isomers have different physicochemical characteristics for example pKa, the same problem cover diastereoisomers.
Enantioselective PK may affect absorption:
D-dopa is less absorbed than L-dopa
D-methotrexate is less absorbed than L-methotrexate
cis - lycopene is absorbed to a greater extent than trans - lycopene
L - cephalexin inhibits the absorption of D - cephalexin from the gut
Enantioselective PK may affect distribution:
  • impact on affinity to blood proteins
  • impact on affinity to transporter systems
  • competition between different form
  • different redistribution with bille depending on racemate
R-methadone has a greater volume of distribution than S-methadone
R - (-) - disopyramide binds less to blood proteins than S - (+) - disopyramide
Cis-cis - mivacurium has a larger volume of distribution than cis-trans, trans-trans
S - propranolol binds more strongly (competes) with plasma proteins than R - propranolol
R - sulbenicillin binds to plasma proteins weaker than S - sulbenicillin
R - latamoxsef binds to plasma proteins more strongly than S - latamoxsef
R-carbenicillin binds to plasma proteins more strongly than S-carbenicillin
Metabolism & Enantioselective PK:
R - propafenone delays the metabolism of S - propafenone
S - nitrendipine is an inhibitor of R - nitrendipine metabolism
R - verapamil is less affected by the first-transition effect than S - verapamil
R - ketoprofen in most species is transformed into S ketoprofen (rodent, dog, monkey, horse, cat) the Asian elephant is the only species that converts S into R
A very interesting case is a chiral inversion in the liver (see examples below)
flobufen (Skalova L. et al 2001)
ibuprofen (Doki K. et al 2003)
pranoprofen (Imai T. et al. 2003)
ketoprofen (Lees P. et al. 2003)
fenoprofen (San Martin M.F. et al. 2002)
albendazole (Virkel G. et al 2002)
thalidomide (Erikson T. et al. 2001)
clopidogrel (Reist M. et al. 2000)
D-leucine (Hasegava H. et al. 2000)
thiaprofenic acid (Erb K. et al 1999)
pantoprazole (Masubuchi N. et al. 1998)
styripentol (Tang C. et al. 1994)
lifibrol (Walters R.R. et al. 1994)
Tolperizon (Yokoyama T. et al. 1992)
Stereoselective elimination:
R-methadone has a longer half-life than S-methadone
R-ibuprofen has a shorter half-life than S-ibuprofen
(-) - mefloquine has a longer half-life than (+) - mefloquine
R - (-) - ketamine inhibits the elimination of S - (+) - ketamine
(+) - terbutaline inhibits tubular reabsorption of (-) - terbutaline
R - sotalolol reduces the renal clearance of S - sotalolol
R - flurbiprofen is excreted from the bile to a greater extent than S - flurbiprofen
R - carprofen is secreted from the bile to a greater extent than S - carprofen
R - sulbenicillin has a lower renal clearance than S - sulbenicillin
R - (+) - propranolol is eliminated more slowly than S - (+) - propranolol
Z - doxepin has a stronger antidepressant effect than E - doxepin
Pharmacodynamics:
Z - doxepin has a stronger antidepressant effect than E - doxepin
S (-) bupivacaine is less toxic than racemic bupivacaine
Of course, it's only the peak of the iceberg so, please check:
.... now > 1000 results ....
examples with references are available in my free e-book (polish version only)
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
5 answers
Currently working on a new drug, previously tested on cell culture studies only. Will administer through IP injection. How do I assess its permeability into the brain and across the bbb? Thanks.
Relevant answer
Answer
In addition to the answer of Geertje you should carefully purge the vascular system by perfusion of saline or an appropriate buffer
best regards , Gert
  • asked a question related to Pharmacokinetics
Question
1 answer
I have to study the pharmacokinetics of floating sustained-release tablets in rats. I have read about the research article regarding the same. But they did not mention clearly, what type of assembly was used for the dosing of the mini-tablets (unbreakable) in rats. I am looking for a clear idea regarding the assemble and procedure of dosing used during the study.
Thank you in advance.
Relevant answer
Answer
You can use the device for PCcaps Capsules, which is designed for rats.
  • asked a question related to Pharmacokinetics
Question
3 answers
I came across with this study here, https://www.clinexprheumatol.org/abstract.asp?a=5066
And this study says: "The sample size calculation was done to compare two different MTX starting dose regimens, based on a study investigating the pharmacokinetics of 25 mg of methotrexate (22) assuming a 67% higher MTX level in the accelerated dose regime and a dose-proportional bioavailability. We calculated that 9 patients in each dosing group will be sufficient to detect the expected difference in the pharmacokinetics of methotrexate (α=0.05, β=0.80)."
Any idea of what formula that they are using to come up with the 9 sample size?
Relevant answer
Answer
it depends on your hypothesis, how big do you anticipate a difference will be ? this difference should at best be of clinical relevance, so that if the study is negative, that we have learned that this covariate is not of clinical relevance ?
  • asked a question related to Pharmacokinetics
Question
7 answers
I want to establish the correlation between in vitro dissolution profile with in vivo pharmackinetic profile. Can any one please tell me how i can establish in vitro in vivo correlation (IVIVC)?
Relevant answer
Answer
Many thanks @ Simon AA Davis
  • asked a question related to Pharmacokinetics
Question
1 answer
I have in vitro dissolution data for generic drug and I want to predict in vivo pharmacokinetics parameters, please suggest me free software tools or high quality R packages that can achieve this. Also please share suitable keywords I can use to search on this topic other than In- Vitro In Vivo Correlation (IVIVC)!
Relevant answer
Answer
Hello,
The best software isn't free. It's expensive. You can try the PKMP trial version https://aplanalyst.com.
  • asked a question related to Pharmacokinetics
Question
2 answers
Can Microsoft Excel be used instead of the popular NONMEM for population pharmacokinetics?
Relevant answer
Answer
Hello Anna,
Lots of thanks for the response and the link was very helpful.
I am grateful indeed.
  • asked a question related to Pharmacokinetics
Question
4 answers
When I viewed the PK parameters of both warfarin and candesartan, I found that both medications have 99% plasma protein binding. I then went to check and confirm there will be PK interaction through resources and I was surprised when I found no interaction! So can it be? Any explanation?
Relevant answer
Answer
Hi
Conclusion about no interactions probably cover lack of meaningful PD observations see:
" Candesartan cilexetil produced a 7% decrease in trough plasma warfarin concentration but this had no effect on prothrombin time. "
So in some cases pharmacokinetic interaction can exist but have no impact on clinical findings. Probably this is why you find conclusions about lack of interaction. Finally protein binding in blood plasma not must be most important aspect of interactions between both drugs
from another hand:
" Candesartan had no effect on S-warfarin 7-hydroxylation. In contrast, S-warfarin inhibited candesartan metabolism by the wild-type (K = 17microM) greater than by the Leu359 variant (Ki = 36 microM). These findings suggest that CYP2C9*3 may change not only the metabolic activity but also the inhibitory susceptibility compared with CYP2C9*1. "
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
5 answers
Is there any reference/Book/Software that shows only the pharmacokinetic parameters of all medications available? For example, if I want to know the absorption, metabolism of acetaminophen, I should open the monograph in this reference and see all the information I need, the same goes for omeprazole, cetirizine, any medication. Is there such a resource/reference?
Relevant answer
Answer
Hi
To be onest from my point of view best are FDA documents related to specific drug submission its better than any database (for example drugbank https://go.drugbank.com/drugs/DB00341) ....
for example
google, key words: fda cetirizine pharmacokinetics accedata
Or maybe such books will be better
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
3 answers
Hello,
I would like to plan a treatment regime with subcutaneous administrations, but I have only spare pharmacokinetic literature data on the compound I use in my experiments. Most of the data are from IV administrations, and I am wondering, which of the followin parameters can I use to estimate pharmacokinetics of the same compound after subcutaneous administration?
- volume of distribution
- clearance
- elimination rate constant and half-life
Thanks!
Relevant answer
Answer
Hi,
You can't predict or simulate S.C. pharmacokinetics without knowledge about bioavailability after S.C. administration in vivo. After you have this information you can simulate or calculate „treatment” – multiple-dose experiments. If you have no flip-flop effects or no issues with non-linear PK then sampling intervals after S.C. should be more less in the same range (zero-last sampling point) as in IV. So planing S.C. should not be very complicated. Of course, you should use some let say general rules for sampling optimization in singe dose studies. See the table 2 from my paper below. Should be useful.
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
6 answers
  • What is the time taken for steady state to be established in adults for ( Mycophenolate mofetil ) ?
Relevant answer
Answer
Please bear in mind that there is a large interindividual variability in the bioavailability and therefore the plasma concentration of parent mycophenolate and active metabolite. The variation can b as large as 30%. You may consider checking the plasma concentration in critical cases.
  • asked a question related to Pharmacokinetics
Question
11 answers
how can i collect many blood sample from the rat for pharmacokinetic study without killing the rat??
Relevant answer
Answer
Hi
Please try sparse sampling method. For example for one time point you can take single sample from 1-3 different rats (rat number 1, 2, 3) for next point single sample from 3 next rats (rat number 4, 5, 6). You can reduce number of animals and leave them alive.
Examples how software can solve such set of data in NCA PK:
Example of study design:
I hope this helps you .... and your rats as well :-)
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
5 answers
I am interested in soaking a drug in a gel foam and then applying the foam topically in vivo to try to obtain a more sustained release of the compound. Do you have any experience with this please? Pros / Cons?
Relevant answer
Answer
Yes. I did.
  • asked a question related to Pharmacokinetics
Question
7 answers
Is it necessary to construct the calibration curve of the drug in plasma for in-vivo pharmacokinetic study? Can we use simple calibration curve of drug constructed in solvent not in the plasma matrix, for quantification of the drug in plasma samples?
Relevant answer
To establish the calibration curve in a matrix similar to your unknown samples, in this case your matrix is plasma, is considered the best option to account for matrix interference with your analytes of interest. It is known as "Matrix-Matched Calibration". It is a common practice when you expect an intereference that may decrease or increase the analytes' signal in the Mass Spectrometer.
To establish the calibration curve in a matrix similar to your unknown samples, in this case your matrix is plasma, is considered the best option to account for matrix interference with your analytes of interest. It is known as "Matrix-Matched Calibration". It is a common practice when you expect an interference that may decrease or increase the analytes' signal in the Mass Spectrometer.
Thus, to answer your question in simple words, yes it is necessary, at least from analytical point of view.
You may refer to some publications about matrix effect in mass spectrometry, like on of my papers in my profile.
  • asked a question related to Pharmacokinetics
Question
8 answers
If comparing formulations of curcumin reported in the literature, which are also available commercially, by their Cmax values, if they have all used different doses, is it scientifically appropriate to dose-normalise each Cmax to say 100mg and then compare their bioavailability that way?
Could you compare liposomes with micelles in this way, native to micelles etc
  • asked a question related to Pharmacokinetics
Question
4 answers
Please suggest the name of software freely available online for calculation of pharmacokinetic parameters
Relevant answer
Answer
As previously mentioned by Siddhartha Dutta
I proposed that the http://www.swissadme.ch/ is the best choice
All the best
  • asked a question related to Pharmacokinetics
Question
7 answers
I found in some of the articles that the Cmax and Tmax of Test and standard to be the same in the pharmacokinetic study but in our experiment both Cmax and Tmax of test were found to be increased as compared to standard. Is our result ok????Please suggest.
Relevant answer
Answer
Por su puesto esto depende de la Biodisponibilidad de cada sustancia y las condiciones de su administración y eliminación. Lo más importante es como afecta la Seguridad y la Eficacia en Terapéutica
  • asked a question related to Pharmacokinetics
Question
3 answers
I am interested on the general opinion regarding how transferrable/comparable are the Km/Vmax values for membrane transporters obtained for example in vitro using transfected cells and ex vivo (such as isolated membranes) or in situ (perfusion studies).
If anybody has a good paper to send me on the topic, I'd really appreciate it.
Thank you in advance!
Relevant answer
Answer
Michaelis–Menten kinetics is one of the best known models of enzyme kinetics in in vitro drug elimination or drug-drug interaction experiments. it consists of two parameters, the maximum reaction rate (Vmax) and the Michaelis constant (Km) describing the rate of enzymatic reactions by relating reaction rate (V) to the concentration of a substrate ([S]). By definition, Km is equal to the concentration of the substrate at half Vmax. It can also be thought of as a measure of how well a substrate complexes with a given enzyme, otherwise known as its binding affinity. An equation with a low Km value indicates a large binding affinity, as the reaction will approach Vmax more rapidly.
  • asked a question related to Pharmacokinetics
Question
3 answers
We are currently trying to create a hepatocyte clearance method that can be used for compounds which are slowly cleared. To do this we are plating the hepatocytes on collagen 48-well plates (viability prior to plating determined using trypan blue). We allow the cells to adhere for 6 hours, change the media, leave the plates overnight and then the next day carry on with metabolic competency studies over a 24 hour period. The hepatocytes we use are these and we follow the supplier instructions: https://www.thermofisher.com/order/catalog/product/WICP10?SID=srch-srp-WICP10#/WICP10?SID=srch-srp-WICP10
While we are carrying out the metabolic competency studies we would like to determine how viable the cells are over the 24 hour period. We initially thought to use CellTiter Glo however, it doesn't seem as though it is lysing all of the cells in the well. We use CellTiter Glo according the the suppliers instructions - take the plate out allow it to equilibrate for 30 minutes at room temperature, add the CellTiter Glo in a 1:1 ratio and shake the plate at 90RPM for 30 minutes before measuring luminescence.
We were wondering if we could maybe add an additional lysis buffer alongside CellTiter Glo? To see whether that would help in fully lysing the cells over the 30 minute period. I've also read some papers online which talk about measuring Caspase 3 activity or Lactate Dehydrogenase... Wondered if anyone could shed some light?
I am a new researcher so please excuse my lack of experience..
Thank you :)
Relevant answer
Answer
Go forAlamar Blue assay that does not require lysis at all.
  • asked a question related to Pharmacokinetics
Question
5 answers
I would like to do some transport assays using tissue from mice that I have experimentally perturbed
Relevant answer
Answer
The publications of Peter Meier describing isolation of hepatic membrane vesicles may be helpful. e.g. J Cell Biol. 1984 Mar;98(3):991-1000.
  • asked a question related to Pharmacokinetics
Question
4 answers
HI,
Trying to engage the wits of my fellow compatriots on this platform to seek online resources that provides free learning opportunities on fundamentals for PK. Looking for something like coursera that can be a self-learning source providing beginner to advanced training. Should be free!
Thanks,
Andy
Relevant answer
Answer
Hi Andy,
Link works correctly but if not in your PC then try with phrase "boomer pk course" in google.com and click first link from the top,
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
4 answers
I have the invitro and cell based invitro concentration of a molecule . But I am struggling with deciding the concentrations that should be screened in mice for the pharmacokinetic study of the molecule. How can we extrapolate the invitro data for setting up invivo experiments?
Relevant answer
Answer
Dear Tejosmita Sen
Few points should be useful I hope:
- develop bioanalytical method for your molecule determination in blood plasma (or blood plasma and whole blood after hemolysis) without a method you cant do your PK study. Moreover you cant make next steps.
- check ex vivo using blank full blood cell fraction and blood plasma, or hemolysed full blood (frozen blood) does your molecule transfer to RBC or not (then you will know what analytical matrix will be useful for you.)
- Calculate LogP of your molecule. If its lipophilic structure with LogP more less >1 then please remember how big impact it could have for binding with plasma proteins (recovery of your analytical method), affinity for other proteins for example with enzyme activity (CYP) (first pass effect possible).
- decide about route of administration and local toxicity (local toxicity should be a part other tox studies) If IV is possible its easier route for you, if not IP could be usefull. Please remember after IV take first samples immediately after drug administration for example 30 sec, after drug administration 2 min 5 min 15 min 30 min ...... its most important samples because show close to t0 observed concentrations which can go dawn with very high rate.
- use tox studies for planning dose which could be well tolerated
- check carefully what is available in relation to monologues (NCBI etc.)
- make pilot study using 3 animals and for example three doses. Example 1mg/kg (one animal), 3 mg/kg (one animal), 9 mg/kg (one animal). If its IV from fist take sample after 30 sec, from next after 30 sec and from last after 3 hours. Analyse data and trends for first PK study sampling optimization.
- With first PK mice study my suggestion is applying sparse sampling. and two stage approach if sampling will be to short to cover whole PK.
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
5 answers
I am aware this is a rather odd question but I couldn’t find any answers to this in the literature. One study has used sperm cells as a drug delivery tool (see below) but I was wondering if it might have broader implications. Would sperm injected into the bloodstream survive for long? What about if it was taken orally? Would appreciate any thoughts!
Relevant answer
Answer
Sperm is typically prevented from exposure to blood - because even a male's own sperm is seen by their immune system as "foreign", leading to the formation of anti-sperm antibodies. This can be a problem in clinical situations such as after testicular trauma, leading to infertility.
As such, I dont think sperm delivery into the bloodstream is practical and although I dont have any specific data on it, I believe the survival of sperm in the blood stream would not be very long. The article you have linked to seems to be suggesting the utility of a sperm delivery system for anti-tumour therapy into the female genital tract (in the lumen, not via the bloodstream) to treat malignancies like endometrial cancer. I couldnt access the full text, so not sure how rigiriusly they have tested this - certainly sounds like an interesting idea!
  • asked a question related to Pharmacokinetics
Question
3 answers
There are numerous articles about development of various chitosan-based nanoparticles for parenteral drug/gene/vaccine delivery but I was unsuccessful to find any references about the pharmacokinetics of these nanoparticles. Are they degradable inside cell endosomes? Could such nanoparticles ever be eliminated from the organism after parenteral administration?
Relevant answer
Answer
Dear Georgi,
Looking in vitro studies looks that chitosan persist long time and could pass from mother to daughter cells through several mitotic cycles.
" Many polymers (especially the chitosan) and their nano/microparticles, commonly referred to as biodegradable/biocompatible, have been investigated for drug delivery and other medical purposes; however, the mechanism of their degradation has not been fully elucidated in biological systems. Evidence for their in vitro or in vivo degradation is very limited or conflicting. The FDA has approved some polymers for drug delivery and certain medical applications [5] because their degradation products are deemed to be biocompatible and eliminated from the body. However, once these polymers have formed nanoparticles by crosslinking (either by covalent, ionic or other bonds), it is not known whether those nano/microparticles are entirely biodegradable or biocompatible. "
source:
It looks like we currently don't have enough data to say that chitosan doesn't have deep compartments. If it is transmitted from cell to cell in mitotic cycles, this indicates strong binding to cell structures and a very low level of elimination. It is very likely that some fraction of chitosan may be eliminated very slowly and stay in the body for a long time. Chitosan with more 73% degree of deacetylation is hardly degraded. Highly deacetylated chitosan (over 85%) shows a low degradation index in the aqueous environment and will degrade after a few months. This means that the kinetics of elimination of this fraction is non-linear. It also requires the creation of more complex pharmacokinetics models. Until now such models probably are not described in literature.
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
7 answers
I am working on pharmacokinetic studies , and trying to find out the plasma drug concentration and facing trouble in this . can anyone help me how you estimate drug concentration from plasma??
Relevant answer
I believe you need to change some of the solvents used, as as Elena V Romanova said, their RT may be the same or similar to your analyte.
Best of luck.
  • asked a question related to Pharmacokinetics
Question
3 answers
What's the difference between food and drug kinetics in human body?
Relevant answer
Answer
Dear Naïrouz Benzeggouta
If I correctly understand your question is not about food impact on drug PK but exactly "pharmacokinetics" of food components. Its very interesting question. Of course term "pharmaco" could be used only for drugs not for other substances.
In general food contents take part exactly the same process like drug after per os administration :
- liberation
- absorption
- distribution
- redistribution
- metabolism
- elimination
Food intake is continuous process, that means substances available in the food are absorbed regularly after meal. In such situation we can talk or describe in scientific manner "kinetics" of some food components (single well defined substance) but not "food pharmacokinetics".
In case of food components is very complicated because many components of food are absorbed in very composite environment from chemical and physiological point of view. Food per se change pH, motility, blood perfusion, and other features of the stomach. That change cover not only stomach but bile ejection and other process influencing absorption every single substance - content of the food. Finally one single meal could deliver millions different substances into gastro-intestinal tract (GIT) which interact between each other in absorption phase and other kinetic (not pharmacokinetic) phases .....
Concluding your question there are many differences between
- drug pharmacokinetics (if we talk about drug)
- food components kinetics (if we talk about any content without PD activity)
But in both cases we can explain mechanisms and processes using current tools proposed by pharmacometrics.
Hope it helps
Best regards
Tomasz
  • asked a question related to Pharmacokinetics
Question
7 answers
I'd like to estimate drug plasma concentrations at a specific time. All participants were patients using a benzodiazepine chronically. The equation I want to use is included in the attachment, but whenever I fill it out I end up with ridiculously high plasma concentrations. I was hoping someone could show me how to fill in the equation using the example case below:
Body weight= 60kg; Temazepam 20mg oral administration of hard capsule every 24 hours; Last administration is 3 hours before testing; F=0.96; Vd= 1.4L/kg; Cl= 1.19L/min/kg; Ka= 0.35; Kel= 0.05
Relevant answer
Answer
Hi Frederick Vinckenbosch , yes, you're right. By solving the equation you get 0.25084 mg/l (by using the original units: 20mg and 84L (from 1.4 L/kg*60kg)) which equals 0.25084 µg/ml or 250.84 ng/ml.
Problem solved, then!
Regards, Luis
  • asked a question related to Pharmacokinetics
Question
4 answers
I've conducted a trial where I have administered a drug through several routes, including IV. I have plasma concentrations at various time-points.
Now I'm trying to find a program that can help me calculate the half-life of elimination and absorption, and the bioavailability. Any suggestions as to what software to use?
Relevant answer
Answer
  • asked a question related to Pharmacokinetics
Question
6 answers
I have the values of all pharmacokinetics parameters for both the reference and test formulations. How can I calculate the effects of formulation, period, sequence and subjects from these parameters?
Relevant answer
Answer
I have found this tool in the web, I think that it uses the IC of 90% and acceptance criteria of 80-125. Please check.
  • asked a question related to Pharmacokinetics
Question
1 answer
I injected two injections into rats via tail vein, irinotecan injection and an herbal extract injection-A (composed of Astragalus membranaceus extract, ginseng extract and oxymatrine). Group1 received only irinotecan; Group2 received a mixture of irinotecan and A; Group3 received A first, and irinotecan was injected 15 minutes later. The injection volume of each group and the concentration of irinotecan in each group were the same. The data demonstrated that at 5 minutes after injection, there was no difference in the plasma concentration of irinotecan between group 3 and group 1, while the plasma concentration of group 2 was increased about 5 times compared to group 1, and AUC was also significantly increased. I analyzed the concentration of irinotecan before and after mixing of the two injections in vitro, no difference. The change in pH after mixing also did not cause too much conversion of lactone and carboxylate forms of irinotecan (lactone forms still accounts for the majority). So why did the injection of a mixture of irinotecan and another drug increase the rat plasma concentration of irinotecan?
Relevant answer
Answer
Hello Yanfei, it is highly probable that your extract inhibited the metabolizing enzymes for irinotecan. Co-administration of certain agents either induces the metabolizing enzymes (leading to rapid clearance/elimination, decreased duration of action and perhaps therapeutic failure) or inhibits the said enzymes, leading to slow clearance/elimination, increased plasma concentration, increased duration of action and perhaps toxicity.
Was this helpful?
  • asked a question related to Pharmacokinetics
Question
3 answers
Is there any pharmacokinetics data for metoprolol tartrate (Cmax) and its solubility?
Relevant answer
Answer
You can go through these links:
Scale-up effects on dissolution and bioavailability of propranolol hydrochloride and metoprolol tartrate tablet formulations.
  • asked a question related to Pharmacokinetics
Question
5 answers
Hi,
I want pharmacokinetic data (Plasma conc. vs time) of certain drugs to build physiologically based pharmacokinetics models. How can I find such data for the studies listed in clinicaltrials.gov? In the results sections, only the pharmacokinetic parameters extracted from that data is given.
Relevant answer
Answer
Thank you, Dr. Tomasz and Dr. Patrice.