Science topic
Drug Delivery - Science topic
Pharmaceutical Drug Delivery
Questions related to Drug Delivery
This question investigates the potential of using specially designed DNA molecules to improve the precision and effectiveness of drug delivery in cancer treatment.
Some of them did the drug release study by Franz diffusion cell . In some other papers people done drug release study by swelling the material into the saline and recording the readings in particular time intervals. But both are completely different environment. Which is correct method to evaluate the drug release for wound healing application.
kindly share your opinion. Thank you
I am working on drug delivery systems, Are there any tests, or criteria to evaluate the drug delivery systems
I saw a lot of research showing the application of DNA as starting material for synthesis of patterned DNA hydrogels or nucleospheres that utilize the self-complementarity of custom designed synthetic oligonucleotides to form such nano and microstructures (1). Another technology exists which uses DNA along with metal ions to form particles through coordination driven self assembly (2). Researchers have also shown the application of such DNA origami structures constructed from custom oligonucleotides and phage DNA in orthotopic tumor-bearing mice models for efficient drug delivery (3).
I am curious to know if we can isolate DNA from other cells (Plants/bacteria/mammalian) and use it to generate such DNA micro or nanoparticles that could have drug delivery applications. What would be the limiting factors in this ?
1.
2. Engineering multifunctional DNA hybrid nanospheres through coordination-driven self-assembly. https://doi.org/10.1002/anie.201810735
Please provide a detailed explanation of the process, mechanism, and structural changes involved in water-based gels controlling the release of drugs.
I am working on a drug delivery, I have to study the nano drug carriers, As of now I am reading from research papers. Are there any databases?, so I can cover all the existing nano drug carriers. It will help me to study them effectively.
- How we can use Resolvability parameters in pharmaceutical sciences. Like in drug delivery, or others
I am currently working on a project involving liposomes and need to determine the maximum volume of siRNA that can be added to a 2.5 mL liposome solution with a total lipid concentration of 10 mg/mL.
I need advice on how to calculate or estimate this volume.
Hello,
I want to load Remedesivir (RNA polymerase inhibitor) into mesoporous nanoparticles (MSNs). The issue is drug is temperature and pH sensitive and it degrades quickly. I need advice on its loading.
1- Should it be loaded to at the last step of MSN synthesis. Problem with this MSNs are extracted later on through reflux and 60 degree temperature is required. It degrades drug.
2- Should it be loaded after extraction making an ice bath but how long it should be kept on stirring?
3- Later I want to coat it with chitosan MSN loaded Remedesivir and then coating with chitosan. I am concerned about leaching and degradation of drug. Should i load drug first and then add drug?
Thank you for your suggestions.
If you prepare drug-loaded nanoparticles but only obtain microparticles (~2,000-4,000 nm), PDI~0.2, how can you reduce the size to reach ~100 nm?
This question addresses the ongoing challenge of improving cancer therapies by focusing on the development of advanced drug delivery systems that can more accurately target cancer cells, thereby reducing side effects and increasing the effectiveness of treatment. Nanotechnology offers a promising avenue for this research due to its potential to create highly specific, controlled, and responsive drug delivery platforms.
I have prepared some polymeric microspheres from chitosan and PCL. However while performing in vitro degradation study, I found difficulties in both above mentioned methods. Here I am mentioning one-by-one possible methods and their drawback I faced and will appreciate if anyone can give a better and doable solution for the below-mentioned methods?
1) By change in weight of microsphere:
Literature has told to take a certain amount of polymeric microspheres in a tube and incubate with different physiological solutions for a pre-determined period. Then to remove the microspheres from the tube, wash it with d.H2O and re-weigh it. Then to calculate the % of weight loss
(degradation)= Initial weight (W0)- weight at that time (Wt)/W0 X 100
The problem I faced: While washing the microspheres with water at a pre-determined period and centrifugation to settle all the microspheres as pellet and other buffers and water as supernatant, there is a chance of microsphere loss during discarding the supernatant of the washing step. You can't be careful enough because PCL is a hydrophobic polymer and it won't form sticky clot at the bottom of centrifuge tube after coming in contact with water. So, when the last portion of the supernatant is about to leave the tube mouth, it will carry some microspheres with it. So, thereby reducing the original weight of the test sample. So there is no meaning of doing a degradation study if I already got the Wt<W0 because of sample loss.
P.S: I am talking about microspheres whose size range from 50-300 um, just like wheat flour. So don't expect me to visibly avoid sample loss during the discarding process because it's a microsphere, not a bead.
2) By measuring the diameter of PCL microsphere from microscopic image processing software.
I tried another method for doing a degradation study by visible parameters like size. After incubation of a certain weight (mg) of microspheres in 3 different PH of PBS for a predetermined period followed by taking 100 ul of the suspension on a slide and observing under 40X microscope and measuring the change in size of the microsphere by image processing software. However, I didn't get any significant change in microsphere size even after maintaining a narrow range of size distribution while choosing microspheres of an image for analysis.
Apart from that, I didn't observe any swelling of the microsphere i.e. increase in microsphere area or diameter on Day 1, rather a significant decrease in average area and diameter. Usually, in case of hydrophilic polymer incubation in PBS, we are expected to observe initial swelling because of water absorption followed by hydrolysis of the polymeric backbone, which results in size decrease of polymeric microspheres.
The problem I faced: There is no initial swelling or significant degradation of microspheres throughout the study period. Only the change in the spherical shape of microsphere was observed after day5 of incubation in PBS and significant degradation and fungal growth observed around day45.
Can anyone suggest me another doable methodology for checking degradation of PCL microsphere ranging from 30-200 um.
I want to develop a nanoparticle-based drug delivery platform for burst/instant release of any drug. Kindly suggest ways to do that? What type of release mechanism can be adopted?
Usually, applied drug release mechanisms like pH-sensitive drug delivery works in a sustained release manner, however, I am looking for techniques (external stimuli) that can promote instant release of drug from the carrier. Thanks in advance for your reply.
Hi
I am working on liposome formulation. we are trying for reproduction of the size at range of +or- 20 nm. but Malvern zeta Sizer always showing different size with 2-3 peaks always.
There are number of publications just mentioned the size in number like ZXC +/- AB.
really frustrated with this first problem
and the second problem is losing hope to trust literature publications.
please share your experience, guide me if possible. help me please.
Thank you.
JR
I am looking for a plain/empty gel system, which we can directly buy and load the drug for the purpose of transdermal delivery. Your kind help will be appreciated.
Hello. Recently, I want to inject the adenosine in vivo to see its effect on the behaviors. But I am not sure whether it can cross blood-brain barrier. If it could, I would choose easier drug-delivery way such as i.p. injection. Could any expert give some advises? Thank you.
I am working on a Speiolite clay-based nanosystem for drug delivery. In order to check the Particle size and zeta potential of the systems what would be the ideal medium to disperse the nanomaterial?
TIA
Please feel free to ask if you need more information
We synthesized a core-shell magneto-electric nanoparticles for drug delivery application, required layer-by-layer deposition method for the synthesis. How it is employed? What's the exact procedure? Also what will be the expected size range of core and shell?
Anyone Can suggest me Lignin Bio-polymer application and its uses or any research which is currently happening in Lignin Nano-particles or lignin nano-composites materials in human welfare and in drug delivery.
Write a short summary of 250 words on why mimicking physiological conditions or clinical situation important for translational research. (Free ChatGPT (July 13 2023 around 1100am JST)
Answer from ChatGPT.
Mimicking physiological conditions or clinical situations is of paramount importance in translational research due to its ability to bridge the gap between laboratory findings and real-world applications. Translational research aims to translate scientific discoveries and knowledge into practical solutions that benefit human health. By replicating the complex and dynamic environments found within the human body, researchers can gain valuable insights into the efficacy, safety, and mechanisms of potential interventions.
One key reason for mimicking physiological conditions is to enhance the relevance and reliability of preclinical studies. Human physiology is intricate, and diseases often manifest within a specific physiological context. By recreating these conditions in experimental models, researchers can better understand how interventions may interact with the body and predict their potential effects on human patients. This approach helps to minimize the risk of false positives or false negatives, which can occur when experimental conditions do not accurately represent the clinical reality.
Furthermore, mimicking physiological conditions enables researchers to investigate the intricate interplay between different biological systems. The human body operates as a complex network of organs, tissues, and cells, all interconnected and influencing one another. By recreating this complexity, researchers can study the holistic effects of interventions, uncover unexpected side effects, and identify potential mechanisms underlying observed outcomes.
Additionally, mimicking clinical situations allows for the evaluation of interventions under realistic scenarios. For example, testing drugs or therapies in animal models that mimic the disease pathology, progression, and response to treatment observed in patients can provide valuable data for predicting their efficacy and safety in humans. This approach aids in the selection and optimization of promising candidates for further clinical trials, ultimately expediting the development of new treatments and therapies.
In conclusion, mimicking physiological conditions or clinical situations is crucial in translational research as it enhances the relevance, reliability, and predictive power of preclinical studies. By closely mirroring the complexities of human physiology and disease, researchers can make more accurate predictions about the potential benefits and risks of interventions, ultimately accelerating the translation of scientific discoveries into clinical applications that improve human health.
References.
Filicori, Marco. "Pulsatile gonadotropin-releasing hormone: clinical applications of a physiologic paradigm." F&S Reports 4.2 (2023): 20-26.
Cahill, Catherine M. "Opioid dose regimen shapes mesolimbic adaptations." Neuropsychopharmacology 45.11 (2020): 1777-1778.
Lefevre, Emilia M., et al. "Interruption of continuous opioid exposure exacerbates drug-evoked adaptations in the mesolimbic dopamine system." Neuropsychopharmacology 45.11 (2020): 1781-1792. https://www.nature.com/articles/s41386-020-0643-x
In relevance to drug delivery, the absorbance of EPIRUBCINE is measured at 480 nm in UV-Spectrophotmeter. Which would be the best selection between UV-Quartz and Glass cuvettes? Is any of this better than the other one or both will give similar results. If anyone can enlighten me on pros and cons of selecting the cuvettes between these two for EPIRUBCINE @480 nm, it would be a great help. Thanks.
Hello,
I'm working on drug delivery in cancer cells. For that, I prepared slides of adherent cancer cells for confocal microscopy. I fixed the cells with 150 ul of 4% paraformaldehyde for 10 min and mounted the cover slip on a glass slide. Then I stored the slides at -20 degC. After one day I did imaging, I found that cells got flattened morphology and some granular structures were seen inside the cells that were totally different from their morphology. Imaging was also not good. I've some doubts regarding this:
1. Whether the incubation time with paraformaldehyde (10 min) was more than required or storage at -20 degC damaged the cells?
2. What should be the optimum time and volume of paraformaldehyde incubation?
3. At what temperature we can store the mounted slides and for how long?
Please guide me regarding this.
Thank you
I am preparing polymeric water in oil (w/o)emulsion where I am using Span-80 as a surfactant. In several papers, it's given that first the emulsion has to be heated around 50-60 degree C followed by keeping the emuslion solution in ice box. By doing this method, I got a microsphere but there is difficulties in capturing the image, which is because of the clumping of microspheres. Later, I read a few Q&A where people mentioned that high temperature is not good for the stability of emulsion as it will degrade the layer of surfactant that is present on top of every microsphere. Usually, it's the surfactant that helps to keep the microspheres away from clumping and keep it separated, which will be actually good for capturing microscopic image.
Now I am confused whether I should heat the emulsion at 50-60 degree C or directly prepare the microsphere, while keeping the emulsion solution in ice box.
Paclitaxel is a hydrophobic drug therefore I dissolved it in DMSO. The issue with DMSO is it has an OD value around 260 nm and the lambda max of paclitaxel is also 263 nm. Stock solution of drug is prepared in DMSO by dissolving 10 mg/mL and further 2 ug/mL of drug dilution was prepared in DMSO as well but OD value is 1.5. I need assistance how to cut the value of DMSO so I can find out the actual peak of drug and then concentration.
Thank you for your help!
With the aim of better loading of gold nanoparticles in drug delivery and cancer treatment.
The goal is to load Doxorubicin on BSA nanoparticles for drug delivery with high affinity.
Hey all
Does chitosan dissolve properly in 1% acetic acid or is it a colloidal solution? I have attached an image for clarity and the source from where I got the protocol.
The following represents the protocol I followed for the synthesis of Chitosan nanoparticle.
1. Weighed 1.25g of Chitosan powder (from Sigma)
2. Made 1% (v/v) acetic acid solution in 500mL double autoclaved water.
3. Added 1-2 mL of tween 20
3. Stirred the solution at room temperature for 24hrs
But the particles are not dissolved and have formed a kind of colloidal solution with froth over the top.
If anyone out there has a standard protocol for making Chitosan nanoparticles, kindly provide it. Also please point out my mistake.
can we formulate nanoparticles with Guar gum and pectin to form a carrier for colon-targeted drug delivery?
How does wide band gap of iron oxide nanoparticles affects drug delivery system and magnetic resonance imaging?
Hi All, I am working with A549 cell line and trying to culture spheroids using low attachment 96 well plates. So far I have attempted some different seeding densities from 2000 to 10,000 cells and can either form very large spheroids (700-900um), which are more compact and have a spherical defined shape, or alternatively smaller spheroids (still fairly big though around 500um) are less compact and not completely spherical. However for my experiment where I wish to add drug compounds (2D IC50 approx 1uM) I am not observing significant size/morphology change on the larger spheroids despite at least a 10uM concentration for 1 week. I am thinking possibly I can try to treat smaller spheroids for a more obvious visual change. Does anyone know how i might successfully make small compact spheroids (less than 500um) which are reproducible with this cell line? Thanks in advance for any help someone may be able to provide.
As hyaluronic acid hydrogels are exteremly hydrophile, is there any possible method to modify them to have hydrophobe hyaluronic acid?
i am currently looking at a nanoparticle formulation for drug delivery to the brain. I want to be able to test the formulation and see if it has any in vitro activity as a drug delivery system
As it is challenging to reach drug into the brain due to BBB (blood-brain barrier) which contain Tight junctions. But other Blood vessels also have tight junction and no perforation. So why BBB is different from other capillaries?
Hello everyone
I Load apolipoprotein on extracellular vesicles (EVs).
After centrifuging, I have EVs, apolipoprotein, and also loaded apolipoprotein. How I can separate them?
And also confirm that?
Thanks for your suggestions.
Hey Guys
Which among the above mentioned is most suitable material for nanoparticle formulation and targeted drug delivery ?
thank you
I want to know any synthetic or natural polymer with same type of acidic conditions solubility of chitosan for drug delivery.
Just like there is a range of zeta potentials that indicates whether Nanoparticles in solution are stable or not, what range of zeta potential qualifies Nanoparticles for use in drug delivery?
I need to try delivering a certain drug to mice's brains via Intracerebroventricular injection. It wasn't done before so I don't know which concentrations and volumes I should try. What is the usual approach for testing drug delivery via ICV?
I want to use silica coated supermagnetic iron nanoparticles loaded with anticancer drug - Doxorubicin for drug delivery. Will simple loading work for this? Or should I do some chemical modifications?
I am using Tween-80 as the surfactant and Glutaraldehyde as a crosslinker for my Ch microsphere formation. Although most have used span-80, few people in research gate also suggested tween-80 can be used. But I want to know what is the concentration.
Also want to know the following parameters:
1) How much con: of Glutaraldehyde will be used as the stock is of 25%? As Some paper has written they used 25% aquous solution of GA, I am confusing how come they use 25% aquous conc: as working where the stock bottle itself written 25%.
2)What is the easy washing method for microspheres after filtering it from the liquid parafilm suspension?
3) What is the optimum drying method for a microsphere? Should I use hot air oven or normal air dry 1st followed by hot air oven.
I'm currently working on exploring the anti-inflammatory effects of a purified plant extract. The compound of interest is a pentacyclic triterpenoid, which is very insoluble in water. This makes drug delivery a bit of a challenge. I'm considering a distilled water/DMSO mixture to act as the vehicle for the compound, which will be administered either through gavage or intraperitoneal injection in mice (C57BL/6). However, since I'm evaluating the anti-inflammatory properties of the compound, I'd like to limit any possible interference from the DMSO, which I know can exhibit anti-inflammatory activity in some doses. Can anyone suggest a possible range of concentrations for DMSO which would not exhibit this anti-inflammatory effect?
We are trying to design Mesoporous Silica Nanoparticles for breast cancer treatment by PhotoDynamic Therapy.
And we are challenging with a question that Which ligand should we add onto the particle for attaching only breast cancer cells when the particles are added into the tumor cells directly by the vaccine (intra-tumoral injection).
Our ligand choice is Anti-HER2 antibody, HA.
We try to research that it is a good choice for us.
#Ligand #HyaluronicAcid #DrugDelivery #BreastCancer #PhotodynamicTherapy #SilicaNanoParticles #Ligand #Antibody #IntraTumoralInjection
Magnetic nanoparticles like ferrites possess self-heating properties and can be used in biomedical applications like magnetic hyperthermia, drug delivery, etc. What are the other applications in which this property can be utilized except biomedical applications?
I have attempted using DMSO as a drug-delivery vehicle for a water-insoluble drug but it tends to precipitate out after injection into mice. I am considering trying this with a NMP/PEG-300 formulation instead but cannot find any literature of this ever having been done before. Anyone have any insight into this method?
Hi.
Is negative zeta potential suitable for liposomes or positive zeta potential?
Some articles have suggested that a negative zeta potential is appropriate for the liposome, and some articles have suggested that a positive zeta potential is a more appropriate factor for liposome delivery of the drug.
Please leave your valuable comments on the effect of zeta potential on drug delivery by liposome
Thank you.
If so, we're hiring! - come and join our team to help develop the next generation of intelligent, wearable drug delivery devices. Research opportunities now available in microsensor integration, transdermal delivery and microfludics, and system electronics/communications. Further details available from Dr Conor O'Mahony - feel free to discuss with us!
The unique physicochemical properties of CNTs make them among prime candidates for numerous applications in biomedical fields including drug delivery, gene therapy, biosensors, and tissue engineering applications.
Carbon nanotubes (CNTs) represent one of the most studied allotropes of carbon. The unique physicochemical properties of CNTs make them among prime candidates for numerous applications in biomedical fields including drug delivery, gene therapy, biosensors, and tissue engineering application
Limitations of carbon dot or carbon dot based products for commercialization.
For a depot injection designed for absorption by macrophage action, which injection route would be optimal to have most drug introduced into the lung by the lymph vessel?
Greeting
I am curious to know whether it is possible to explain and analyze targeted drug delivery methods with the mathematical and physical relationships that govern them, such as the Peppas Higuchi model?
What other models are available in this field?
Why nature-based products are considered biocompatible for drug delivery although many natural products can be toxic or dangerous
I am working on nanoemulsions for topical drug delivery. However, due to limitations with the equipment available, I am wondering is it possible to prepare my nanoemulsion in small volume, and proceed to dilute it with water? Will this process causes breakdown of the micellar structure of NE?
Hope that everyone will be quite fine sir I am working on Self nanoemulsyfying drug delivery of low orally bioavailable drugs but em facing some problem my formulation have zeta potential of -10 and PDI of 0.42 can we consider it as stable and accepted .
Second those who know the concept of self nanoemulsyfying drug delivery system when it taken once directly goes into the gut and form a nanoemulsion due to GIT motality then still the PDI and zeta potential can be considered as standard like conventional nano emulsion because the face problem after dilution with aqous phase and in shelf life ???
Dear all,
Good day.
I'm currently working on a formulation involving an extract from garlic, allicin and will be moving onto the in vitro drug release test. Based on my preparation for the standard curve via UV Vis spectroscopy, allicin is temperature sensitive and has to be placed on ice (the absorbance seems to fluctuate if its left at room temperature over time). The drug release assay seems to be conducted at 37 °C most of the time hence, I'm wondering if it would be a problem if I'm planning to measure the release of allicin from my formulation via UV Vis during the in vitro drug release assay?
Thank you in advance for your time.
What is the best dosage of tetracylcline in vitro effect? i dont know the theurapetic window or index of this drug. please help me guys.
Hello, can anyone please named few potential drug delivery technologies that you think are novel and unique. Our comapny is looking to buy few drug delivery technologies that can serve us to compete for now and few next years. Suggestions are highly appreciated. Thanks in advance.
Regards
For calculation of drug release via Franzcell diffusion, do you consider the whole volume of the media cell? or you just divide the drug concentration by the amount of the initial drug?
Hi,
I wanted to know about the drug release behaviour under HPLC method.
I am trying to get the peak for release profile of nanoparticle-Paclitaxel drug in 1X PBS for different time points whereas I got the peak for loading. Nanoparticle is calcium phosphate. Could anyone suggest the possible solution for the same.
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.
I want to check for apoptosis in the co-delivery system. One of the drugs that are simultaneous with another drug in nanocarrier is prodigiosin. Since the emission wavelength of prodigiosin was similar to that of propidium iodide, necrosis was detected instead of apoptosis. How can I improve my apoptosis results?
Dear all,
Greetings. I purchased MWCNTs, which are >98% carbon basis, O.D. × L 6-13 nm × 2.5-20 μm(Sigma, cat no. 698849) and oxidised them using the method described by Thi Mai Hoa, 2018. A total of 100 mg of pristine MWCNTs is added into 20 mL of 4 M of HNO3 and 10 M of H2SO4 (1:3) and left stirring for 18 h at room temperature after which, it is coated with chitosan in 1% and 2% acetic acid as described by Sobh et al., 2019.
Previous papers have emphasised the importance of the oxidation step to prevent the aggregation of MWCNTs thus, I decided to conduct the dispersion assay as described by Singer et al., 2018 (in their paper,). A total of 20 mg of pristine, oxidised and coated MWCNTs were dispersed in 20 mL of water and sonicated in a waterbath for 30 min followed by an hour of sedimentation. The mixtures were observed over a week. Based on my observation, the pristine MWCNTs sank to the bottom after 24 h. However, the oxidised MWCNTs also appeared to be settling after 48 h (as observed in the picture). The MWCNTs coated with chitosan appeared the same from Day 1 (Picture labelled 1 h).
How important would the oxidation step be in this case? Would it be safe to utilised the coated MWCNTs since it shows no signs of settling? I've attached some pictures from the first hour and a week after sonication. I would appreciate your feedbacks or recommendations. Thank you.
I am searching for better biorobots for targeted drug delivery. Please suggest some microbots/nanorobots which were used in the cancer drug delivery system with working.
Current commercially available implantable pumps are osmotic pumps (www.alzet.com) and programmable micro infusion pumps (www.iprecio.com) in the preclinical/drug discovery market. What would Users like to see in next generation commercially available pumps? (must have, nice to have, short term requirements, long term dream …… in this preclinical/drug discovery market –(non-clinical applications)
For inspiration – commercially available implantable clinical pumps.
I am planning to deliver a drug in C57BL6 mice and the desired concentration is 25mg/kg/day. Assuming that the average weight of the mice is 40 grams and the drug has to be delivered for 2 weeks straight, how much drug do i need to perform the experiment, how to prepare the stock and what kind of osmotic pump should I use?
Hi!
We are using microfluidics to fabricate gel beads. Though they have a central mean, the distribution presents a quite discrete distribution. However for our application we need to strictly rid off bead >60um.
This is not a usual size in cell strainer and I cannot find 60um cell strainer in China. Instead, I ordered some stainless steel net. The problem is that the steel net is quite elastic. Even with pole size to 55±2um, there are still seldom 60-61um beads remain.
Is there any other method to rid off the >60 bead?
Dear Researcher
Respectfully, I am working on drug delivery.
I synthesized Zn-MOF. I am going to load Doxorobicin on it and the study the derivery behaviour.
I tried several times to load. but I did not succeed. Apparently, the drug is not loaded on MOF. Could you Please guide me why not loaded? Theere are several hydroxyl, carbonyl and ether functional groups on the doxorubicin and it seems to be loaded easily. The results indicated no loading on MOF.
Regards
Hi,
I am very new to the study of/working with hydrogels and have not gotten the chance to get into a lab because of covid. So all of my knowledge comes from scientific literature I’ve read over the past 1-2 months, and is very limited. I am trying to make biocompatible microspheres (.5 to 5 microns) that degrade extremely quickly (1-2 minutes ideally). I apologize if my questions are obvious, but I would greatly appreciate any suggestions on how to make this happen. I found several articles cite that pure PEG degrades quickly around 400 degrees, but was wondering if a hydrogel made with PEG would degrade at a lower temp? Would a thermosensitive element to the hydrogel even be necessary at high temps like this, or would complete degradation happen first? What would the timeframe look like for degradation at this temp? I also read a paper that showed the thermal degradation pathways of PEG; is there any concern of accumulation of these products in the body (this one is a major concern, I was thinking a super high water content would help this)? Lastly if used for drug delivery, how fast could complete drug delivery happen if the microspheres were kept dry (with a very small drug and very large pores), once introduced to water and swelled (with or without high temps and heat degradation)?
The most important thing is to end up with biocompatible microspheres that degrade extremely quickly, so it doesn’t even have to happen via heat or with PEG specifically. Any pushes in the right direction (or advice if I have any glaring misunderstandings) would be much appreciated!
Thanks in advance!
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.
Hi to all!
I am currently working on sequential glucose and oxygen delivery in tissue engineering field and I need to choose specific tissue and cells to work on. I wonder that which tissue need the highest amount of glucose during the first days of implantation. is there any valid reference reporting concentration of normal glucose consumption rate in different tissues?
I am currently studying a proteolytically stable peptide that transiently increases blood brain barrier permeability. While the results suggest that it could facilitate drug delivery to the brain, we are interested in assessing the potential downside of such a strategy.
Primarily, we are worried about neuroinflammation. My lab does not have the facilities to properly detect neuroinflammation. Does anyone know of any lab, core facility, or private company that offers services that can determine whether or not the peptide we are testing can lead to neuroinflammation?