Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Pharm Res ; 41(2): 223-234, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38158503

ABSTRACT

PURPOSE: Accurate methods to determine dermal pharmacokinetics are important to increase the rate of clinical success in topical drug development. We investigated in an in vivo pig model whether the unbound drug concentration in the interstitial fluid as determined by dermal open flow microperfusion (dOFM) is a more reliable measure of dermal exposure compared to dermal biopsies for seven prescription or investigational drugs. In addition, we verified standard dOFM measurement using a recirculation approach and compared dosing frequencies (QD versus BID) and dose strengths (high versus low drug concentrations). METHODS: Domestic pigs were topically administered seven different drugs twice daily in two studies. On day 7, drug exposures in the dermis were assessed in two ways: (1) dOFM provided the total and unbound drug concentrations in dermal interstitial fluid, and (2) clean punch biopsies after heat separation provided the total concentrations in the upper and lower dermis. RESULTS: dOFM showed sufficient intra-study precision to distinguish interstitial fluid concentrations between different drugs, dose frequencies and dose strengths, and had good reproducibility between studies. Biopsy concentrations showed much higher and more variable values. Standard dOFM measurements were consistent with values obtained with the recirculation approach. CONCLUSIONS: dOFM pig model is a robust and reproducible method to directly determine topical drug concentration in dermal interstitial fluid. Dermal biopsies were a less reliable measure of dermal exposure due to possible contributions from drug bound to tissue and drug associated with skin appendages.


Subject(s)
Skin , Swine , Animals , Administration, Cutaneous , Reproducibility of Results , Skin/metabolism
2.
Xenobiotica ; 52(8): 770-785, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36314242

ABSTRACT

 The therapeutic concept of antibody drug conjugates (ADCs) is to selectively target tumour cells with small molecule cytotoxic drugs to maximise cell kill benefit and minimise healthy tissue toxicity.An ADC generally consists of an antibody that targets a protein on the surface of tumour cells chemically linked to a warhead small molecule cytotoxic drug.To deliver the warhead to the tumour cell, the antibody must bind to the target protein and in general be internalised into the cell. Following internalisation, the cytotoxic agent can be released in the endosomal or lysosomal compartment (via different mechanisms). Diffusion or transport out of the endosome or lysosome allows the cytotoxic drug to express its cell-killing pharmacology. Alternatively, some ADCs (e.g. EDB-ADCs) rely on extracellular cleavage releasing membrane permeable warheads.One potentially important aspect of the ADC mechanism is the 'bystander effect' whereby the cytotoxic drug released in the targeted cell can diffuse out of that cell and into other (non-target expressing) tumour cells to exert its cytotoxic effect. This is important as solid tumours tend to be heterogeneous and not all cells in a tumour will express the targeted protein.The combination of large and small molecule aspects in an ADC poses significant challenges to the disposition scientist in describing the ADME properties of the entire molecule.This article will review the ADC landscape and the ADME properties of successful ADCs, with the aim of outlining best practice and providing a perspective of how the field can further facilitate the discovery and development of these important therapeutic modalities.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Humans , Antineoplastic Agents/pharmacokinetics , Immunoconjugates/pharmacokinetics , Neoplasms/drug therapy
3.
Regul Toxicol Pharmacol ; 123: 104934, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33872740

ABSTRACT

Systemic toxicity assessments for oral or parenteral drugs often utilize the concentration of drug in plasma to enable safety margin calculations for human risk assessment. For topical drugs, there is no standard method for measuring drug concentrations in the stratum basale of the viable epidermis. This is particularly important since the superficial part of the epidermis, the stratum corneum (SC), is nonviable and where most of a topically applied drug remains, never penetrating deeper into the skin. We investigated the relative concentrations of a prototype kinase inhibitor using punch biopsy, laser capture microdissection, and imaging mass spectrometry methods in the SC, stratum basale, and dermis of minipig skin following topical application as a cream formulation. The results highlight the value of laser capture microdissection and mass spectrometry imaging in quantifying the large difference in drug concentration across the skin and even within the epidermis, and supports use of these methods for threshold-based toxicity risk assessments in specific anatomic locations of the skin, like of the stratum basale.


Subject(s)
Pharmaceutical Preparations/metabolism , Skin Absorption/physiology , Skin/metabolism , Animals , Epidermis , Humans , Mass Spectrometry , Risk Assessment , Swine , Swine, Miniature/physiology
5.
J Am Chem Soc ; 139(9): 3528-3536, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28230359

ABSTRACT

A compact and stable bicyclic bridged ketal was developed as a ligand for the asialoglycoprotein receptor (ASGPR). This compound showed excellent ligand efficiency, and the molecular details of binding were revealed by the first X-ray crystal structures of ligand-bound ASGPR. This analogue was used to make potent di- and trivalent binders of ASGPR. Extensive characterization of the function of these compounds showed rapid ASGPR-dependent cellular uptake in vitro and high levels of liver/plasma selectivity in vivo. Assessment of the biodistribution in rodents of a prototypical Alexa647-labeled trivalent conjugate showed selective hepatocyte targeting with no detectable distribution in nonparenchymal cells. This molecule also exhibited increased ASGPR-directed hepatocellular uptake and prolonged retention compared to a similar GalNAc derived trimer conjugate. Selective release in the liver of a passively permeable small-molecule cargo was achieved by retro-Diels-Alder cleavage of an oxanorbornadiene linkage, presumably upon encountering intracellular thiol. Therefore, the multicomponent construct described here represents a highly efficient delivery vehicle to hepatocytes.


Subject(s)
Asialoglycoprotein Receptor/metabolism , Bridged Bicyclo Compounds/chemistry , Hepatocytes/metabolism , Ketones/chemistry , Liver/metabolism , Polymers/chemistry , Bridged Bicyclo Compounds/metabolism , Crystallography, X-Ray , Drug Carriers/chemistry , Humans , Ketones/metabolism , Liver/cytology , Models, Molecular , Molecular Structure , Polymers/metabolism
7.
Angew Chem Int Ed Engl ; 56(51): 16218-16222, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29073340

ABSTRACT

Targeting of the human ribosome is an unprecedented therapeutic modality with a genome-wide selectivity challenge. A liver-targeted drug candidate is described that inhibits ribosomal synthesis of PCSK9, a lipid regulator considered undruggable by small molecules. Key to the concept was the identification of pharmacologically active zwitterions designed to be retained in the liver. Oral delivery of the poorly permeable zwitterions was achieved by prodrugs susceptible to cleavage by carboxylesterase 1. The synthesis of select tetrazole prodrugs was crucial. A cell-free in vitro translation assay containing human cell lysate and purified target mRNA fused to a reporter was used to identify active zwitterions. In vivo PCSK9 lowering by oral dosing of the candidate prodrug and quantification of the drug fraction delivered to the liver utilizing an oral positron emission tomography 18 F-isotopologue validated our liver-targeting approach.


Subject(s)
Liver/drug effects , PCSK9 Inhibitors , Proprotein Convertase 9/biosynthesis , Small Molecule Libraries/pharmacology , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/enzymology , Liver/metabolism , Molecular Structure , Proprotein Convertase 9/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship
9.
J Pharmacol Exp Ther ; 351(2): 259-69, 2014 11.
Article in English | MEDLINE | ID: mdl-25125580

ABSTRACT

The transient receptor potential (subfamily M, member 8; TRPM8) is a nonselective cation channel localized in primary sensory neurons, and is a candidate for cold thermosensing, mediation of cold pain, and bladder overactivity. Studies with TRPM8 knockout mice and selective TRPM8 channel blockers demonstrate a lack of cold sensitivity and reduced cold pain in various rodent models. Furthermore, TRPM8 blockers significantly lower body temperature. We have identified a moderately potent (IC50 = 103 nM), selective TRPM8 antagonist, PF-05105679 [(R)-3-[(1-(4-fluorophenyl)ethyl)(quinolin-3-ylcarbonyl)amino]methylbenzoic acid]. It demonstrated activity in vivo in the guinea pig bladder ice water and menthol challenge tests with an IC50 of 200 nM and reduced core body temperature in the rat (at concentrations >1219 nM). PF-05105679 was suitable for acute administration to humans and was evaluated for effects on core body temperature and experimentally induced cold pain, using the cold pressor test. Unbound plasma concentrations greater than the IC50 were achieved with 600- and 900-mg doses. The compound displayed a significant inhibition of pain in the cold pressor test, with efficacy equivalent to oxycodone (20 mg) at 1.5 hours postdose. No effect on core body temperature was observed. An unexpected adverse event (hot feeling) was reported, predominantly periorally, in 23 and 36% of volunteers (600- and 900-mg dose, respectively), which in two volunteers was nontolerable. In conclusion, this study supports a role for TRPM8 in acute cold pain signaling at doses that do not cause hypothermia.


Subject(s)
Pain/metabolism , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/metabolism , Animals , Body Temperature/drug effects , Cold Temperature , Cross-Over Studies , Double-Blind Method , Guinea Pigs , HEK293 Cells , Humans , Male , Membrane Transport Modulators/pharmacology , Oxycodone/pharmacology , Pain/drug therapy , Rats , Rats, Wistar
10.
Bioorg Med Chem Lett ; 24(1): 204-8, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24332488

ABSTRACT

HSP90 continues to be a target of interest for neurodegeneration indications. Selective knockdown of the HSP90 cytosolic isoforms α and ß is sufficient to reduce mutant huntingtin protein levels in vitro. Chemotype-dependent binding conformations of HSP90α/ß appear to strongly influence isoform selectivity. The rational design of HSP90α/ß inhibitors selective versus the mitochondrial (TRAP1) and endoplasmic reticulum (GRP94) isoforms offers a potential mitigating strategy for mechanism-based toxicities. Better tolerated HSP90 inhibitors would be attractive for targeting chronic neurodegenerative diseases such as Huntington's disease.


Subject(s)
Drug Design , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Neurodegenerative Diseases/drug therapy , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure , Protein Isoforms/antagonists & inhibitors
11.
Bioorg Med Chem ; 20(1): 498-509, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22100260

ABSTRACT

This paper describes our recent efforts to design and synthesise potent and selective PDE5 inhibitors and the use of in vitro predictors of clearance, absorption and permeability to maximise the potential for dose-proportional pharmacokinetics and good oral bioavailability in man. Optimisation of the preclinical profile resulted in the identification of UK-369003 (19a) and its nomination as a clinical candidate. The clinical pharmacokinetic and safety profile has enabled us to progress the compound to test its efficacy in patients with lower urinary tract symptoms (LUTS) associated with benign prostatic hyperplasia (BPH) and a paper describing its efficacy has recently been published.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/chemistry , Phosphodiesterase 5 Inhibitors/chemistry , Phosphodiesterase 5 Inhibitors/pharmacokinetics , Pyrimidinones/chemistry , Pyrimidinones/pharmacokinetics , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Administration, Oral , Biological Availability , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Humans , Lower Urinary Tract Symptoms/drug therapy , Lower Urinary Tract Symptoms/etiology , Male , Phosphodiesterase 5 Inhibitors/pharmacology , Prostatic Hyperplasia/complications , Pyrimidinones/pharmacology , Sulfonamides/pharmacology
12.
Xenobiotica ; 42(1): 57-74, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21992032

ABSTRACT

PF-184298 ((S)-2,3-dichloro-N-isobutyl-N-pyrrolidin-3-ylbenzamide) and PF-4776548 ((3-(4-fluoro-2-methoxy-benzyl)-7-hydroxy-8,9-dihydro-3H,7H-pyrrolo[2,3-c][1,7]naphthyridin-6-one)) are novel compounds which were selected to progress to human studies. Discordant human pharmacokinetic predictions arose from pre-clinical in vivo studies in rat and dog, and from human in vitro studies, resulting in a clearance prediction range of 3 to >20 mL min⁻¹ kg⁻¹ for PF-184298, and 5 to >20 mL min⁻¹ kg⁻¹ for PF-4776548. A package of work to investigate the discordance for PF-184298 is described. Although ultimately complementary to the human pharmacokinetic data in characterising the disposition of PF-184298 in humans, these data did not provide any further confidence in pharmacokinetic prediction. A fit for purpose human pharmacokinetic study was conducted for each compound, with an oral pharmacologically active dose for PF-184298, and an intravenous and oral microdose for PF-4776548. This provided a relatively low cost, clear decision making approach, resulting in the termination of PF-4776548 and further progression of PF-184298. A retrospective analysis of the data showed that, if the tools had been available at the time, the pharmacokinetics of PF-184298 in human could have been predicted from a population based simulation tool in combination with physicochemical properties and in vitro human intrinsic clearance.


Subject(s)
Anilides/pharmacokinetics , Drug Evaluation, Preclinical/methods , Models, Biological , Naphthyridines/pharmacokinetics , Pyrrolidines/pharmacokinetics , Adult , Anilides/administration & dosage , Animal Testing Alternatives , Animals , Dogs , Drug Discovery , Humans , Male , Microsomes, Liver/metabolism , Naphthyridines/administration & dosage , Pharmacokinetics , Pyrrolidines/administration & dosage , Rats , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/pharmacokinetics , Statistics as Topic , Young Adult
13.
Front Med (Lausanne) ; 9: 931293, 2022.
Article in English | MEDLINE | ID: mdl-35966871

ABSTRACT

Adenosine triphosphate (ATP) released from injured or dying cells is a potent pro-inflammatory "danger" signal. Alkaline phosphatase (AP), an endogenous enzyme that de-phosphorylates extracellular ATP, likely plays an anti-inflammatory role in immune responses. We hypothesized that ilofotase alfa, a human recombinant AP, protects kidneys from ischemia-reperfusion injury (IRI), a model of acute kidney injury (AKI), by metabolizing extracellular ATP to adenosine, which is known to activate adenosine receptors. Ilofotase alfa (iv) with or without ZM241,385 (sc), a selective adenosine A2A receptor (A2AR) antagonist, was administered 1 h before bilateral IRI in WT, A2AR KO (Adora2a-/- ) or CD73-/- mice. In additional studies recombinant alkaline phosphatase was given after IRI. In an AKI-on-chronic kidney disease (CKD) ischemic rat model, ilofotase alfa was given after the three instances of IRI and rats were followed for 56 days. Ilofotase alfa in a dose dependent manner decreased IRI in WT mice, an effect prevented by ZM241,385 and partially prevented in Adora2a-/- mice. Enzymatically inactive ilofotase alfa was not protective. Ilofotase alfa rescued CD73-/- mice, which lack a 5'-ectonucleotidase that dephosphorylates AMP to adenosine; ZM241,385 inhibited that protection. In both rats and mice ilofotase alfa ameliorated IRI when administered after injury, thus providing relevance for therapeutic dosing of ilofotase alfa following established AKI. In an AKI-on-CKD ischemic rat model, ilofotase alfa given after the third instance of IRI reduced injury. These results suggest that ilofotase alfa promotes production of adenosine from liberated ATP in injured kidney tissue, thereby amplifying endogenous mechanisms that can reverse tissue injury, in part through A2AR-and non-A2AR-dependent signaling pathways.

14.
J Med Chem ; 64(1): 326-342, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33356244

ABSTRACT

Sickle cell disease (SCD) is a genetic disorder caused by a single point mutation (ß6 Glu → Val) on the ß-chain of adult hemoglobin (HbA) that results in sickled hemoglobin (HbS). In the deoxygenated state, polymerization of HbS leads to sickling of red blood cells (RBC). Several downstream consequences of polymerization and RBC sickling include vaso-occlusion, hemolytic anemia, and stroke. We report the design of a noncovalent modulator of HbS, clinical candidate PF-07059013 (23). The seminal hit molecule was discovered by virtual screening and confirmed through a series of biochemical and biophysical studies. After a significant optimization effort, we arrived at 23, a compound that specifically binds to Hb with nanomolar affinity and displays strong partitioning into RBCs. In a 2-week multiple dose study using Townes SCD mice, 23 showed a 37.8% (±9.0%) reduction in sickling compared to vehicle treated mice. 23 (PF-07059013) has advanced to phase 1 clinical trials.


Subject(s)
Anemia, Sickle Cell/drug therapy , Hemoglobin A/drug effects , Hemoglobin, Sickle/drug effects , Quinolines/pharmacology , Quinolines/therapeutic use , Animals , Erythrocytes/metabolism , Mice , Oxygen/metabolism , Quinolines/chemistry
15.
J Med Chem ; 63(12): 6423-6435, 2020 06 25.
Article in English | MEDLINE | ID: mdl-31913040

ABSTRACT

The efficacious dose of a drug is perhaps the most holistic metric reflecting its therapeutic potential. Dose is predicted at many stages in drug discovery and development. Prior to the 1990s, dose prediction was limited to the drug "working" at a reasonable dose and dose regimen in an animal model. Through the early 2000s, dose predictions were generated at candidate nomination and then refined during clinical development. Currently, dose predictions can be made early in drug discovery to enable drug design. Dose predictions at this stage can identify critical drug properties for a viable dose regimen and provide clinically relevant context to lead optimization. In this paper, we give an overview of the opportunities and challenges associated with dose prediction for drug design. A number of general considerations, approaches, and case examples are discussed.


Subject(s)
Drug Design , Drug Discovery , Drug Evaluation, Preclinical , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/standards , Animals , Dose-Response Relationship, Drug , Humans , Pharmaceutical Preparations/metabolism
16.
Drug Metab Dispos ; 37(9): 1864-70, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19546239

ABSTRACT

The relationship between rat pharmacokinetics and physicochemical parameters [the partition coefficient between octanol and buffer at pH 7.4 (log D((7.4))) and pK(a)] was studied for a series of tetrahydropyran compounds. Sixteen compounds ranging in log D((7.4)) 0.1 to 1.8 were administered intravenously to rats, and the pharmacokinetic parameters were determined from blood concentration time curves. Across the series, a weak correlation was observed between log D((7.4)) and blood clearance, suggesting that log D((7.4)) values less than 0.5 were required to prevent clearance at hepatic blood flow. In terms of the volume of distribution (V(d)), the compounds fell into three distinct subseries characterized by the number of basic centers and differences in ionization of each basic center at physiological pH. These were referred to as the monobasic, weak second base, and strong second base subseries. All the compounds exhibited V(d) greater than body water, as would be expected from their lipophilic and basic nature. For a given clog P, the strong second base subseries showed higher V(d) than the weak second base subseries, which in turn exhibited higher values than the monobasic subseries. In addition, for the weak second base subseries, V(d) could be tuned by modulating the pK(a) of the second basic center. This relationship was rationalized in respect to the interactions of the ionizable centers with phospholipid heads in the cell membrane and/or lysosomal trapping. Compounds in the weak second base subseries showed optimal V(d), and when combined with a log D((7.4)) of 0.1, driving to moderate blood clearance, one compound showed the optimal pharmacokinetic profile.


Subject(s)
Histamine H3 Antagonists/pharmacokinetics , Octanols/chemistry , Pyrans/pharmacokinetics , Animals , Biological Availability , Buffers , Cytochrome P-450 Enzyme System/metabolism , Half-Life , Histamine H3 Antagonists/blood , Histamine H3 Antagonists/chemistry , Humans , Hydrogen-Ion Concentration , Injections, Intravenous , Kinetics , Male , Microsomes, Liver , Protein Binding , Pyrans/blood , Pyrans/chemistry , Rats , Rats, Sprague-Dawley
17.
J Clin Pharmacol ; 49(5): 513-33, 2009 May.
Article in English | MEDLINE | ID: mdl-19299532

ABSTRACT

Quantitative prediction of human pharmacokinetics is critical in assessing the viability of drug candidates and in determining first-in-human dosing. Numerous prediction methodologies, incorporating both in vitro and preclinical in vivo data, have been developed in recent years, each with advantages and disadvantages. However, the lack of a comprehensive data set, both preclinical and clinical, has limited efforts to evaluate the optimal strategy (or strategies) that results in quantitative predictions of human pharmacokinetics. To address this issue, the authors conducted a retrospective analysis using 50 proprietary compounds for which in vitro, preclinical pharmacokinetic data and oral single-dose human pharmacokinetic data were available. Five predictive strategies, involving either allometry or use of unbound intrinsic clearance from microsomes or hepatocytes, were then compared for their ability to predict human oral clearance, half-life through predictions of systemic clearance, volume of distribution, and bioavailability. Use of a single-species scaling approach with rat, dog, or monkey was as accurate as or more accurate than using multiple-species allometry. For those compounds cleared almost exclusively by P450-mediated pathways, scaling from human liver microsomes was as predictive as single-species scaling of clearance based on data from rat, dog, or monkey. These data suggest that use of predictive methods involving either single-species in vivo data or in vitro human liver microsomes can quantitatively predict human in vivo pharmacokinetics and suggest the possibility of streamlining the predictive methodology through use of a single species or use only of human in vitro microsomal preparations.


Subject(s)
Drug Evaluation, Preclinical/methods , Models, Biological , Pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Biological Availability , Biometry , Half-Life , Hepatocytes/metabolism , Humans , Metabolic Clearance Rate , Microsomes, Liver/metabolism , Reproducibility of Results , Retrospective Studies , Species Specificity , Xenobiotics/pharmacokinetics
18.
J Med Chem ; 62(5): 2245-2255, 2019 Mar 14.
Article in English | MEDLINE | ID: mdl-30281973

ABSTRACT

Due to its implications for both dose level and frequency, clearance rate is one of the most important pharmacokinetic parameters to consider in the design of drug candidates. Clearance can be classified into three general categories, namely, metabolic transformation, renal excretion, and hepatobiliary excretion. Within each category, there are a host of biochemical and physiological mechanisms that ultimately determine the clearance rate. Physiochemical properties are often indicative of the rate-determining mechanism, with lipophilic molecules tending toward metabolism and hydrophilic, polar molecules tending toward passive or active excretion. Optimization of clearance requires recognition of the major clearance mechanisms and use of the most relevant in vitro and in vivo tools to develop structure-clearance relationships. The reliability of methods to detect and predict human clearance varies across mechanisms. While methods for metabolic and passive renal clearance have proven reasonably robust, there is a clear need for better tools to support the optimization of transporter-mediated clearance.


Subject(s)
Drug Design , Metabolic Clearance Rate , Pharmaceutical Preparations/metabolism , Animals , Humans , Hydrophobic and Hydrophilic Interactions , Kidney/metabolism , Liver/metabolism , Membrane Transport Proteins/metabolism , Structure-Activity Relationship
19.
Bioorg Med Chem Lett ; 18(20): 5601-4, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18809326

ABSTRACT

The optimisation of a series of amides for C5a receptor binding and functional activity, and physicochemical properties is described. The initial hit, 1 (IC(50) 1 microM), was discovered during high throughput screening, from which highly potent C5a receptor antagonists (e.g.14, IC(50) 5 nM) were developed.


Subject(s)
Chemistry, Pharmaceutical/methods , Complement C5a/antagonists & inhibitors , Peptides/chemistry , Administration, Oral , Buffers , Complement C5a/chemistry , Drug Design , Humans , Hydrogen-Ion Concentration , Hydrolysis , Inhibitory Concentration 50 , Microsomes, Liver/metabolism , Models, Chemical , Piperidines/chemistry , Protein Binding
20.
Bioorg Med Chem Lett ; 18(20): 5605-8, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18804369

ABSTRACT

Starting from 2, several highly potent C5a receptor antagonists were synthesised through alpha-amide substitution. Attempts to increase the polarity of these compounds through the introduction of basic centres or incorporation into weakly basic heterocycles is described.


Subject(s)
Amides/chemistry , Chemistry, Pharmaceutical/methods , Complement C5a/antagonists & inhibitors , Peptides/chemistry , Administration, Oral , Binding Sites , Complement C5a/chemistry , Drug Design , Humans , Hydrolysis , Inflammation , Inhibitory Concentration 50 , Models, Chemical , Molecular Structure , Piperidines/chemistry , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL