Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
IUCrJ ; 11(Pt 4): 556-569, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38856178

ABSTRACT

Carbonic anhydrase (CA) was among the first proteins whose X-ray crystal structure was solved to atomic resolution. CA proteins have essentially the same fold and similar active centers that differ in only several amino acids. Primary sulfonamides are well defined, strong and specific binders of CA. However, minor variations in chemical structure can significantly alter their binding properties. Over 1000 sulfonamides have been designed, synthesized and evaluated to understand the correlations between the structure and thermodynamics of their binding to the human CA isozyme family. Compound binding was determined by several binding assays: fluorescence-based thermal shift assay, stopped-flow enzyme activity inhibition assay, isothermal titration calorimetry and competition assay for enzyme expressed on cancer cell surfaces. All assays have advantages and limitations but are necessary for deeper characterization of these protein-ligand interactions. Here, the concept and importance of intrinsic binding thermodynamics is emphasized and the role of structure-thermodynamics correlations for the novel inhibitors of CA IX is discussed - an isozyme that is overexpressed in solid hypoxic tumors, and thus these inhibitors may serve as anticancer drugs. The abundant structural and thermodynamic data are assembled into the Protein-Ligand Binding Database to understand general protein-ligand recognition principles that could be used in drug discovery.


Subject(s)
Carbonic Anhydrases , Isoenzymes , Protein Binding , Sulfonamides , Thermodynamics , Humans , Crystallography, X-Ray , Carbonic Anhydrases/metabolism , Carbonic Anhydrases/chemistry , Isoenzymes/metabolism , Isoenzymes/chemistry , Ligands , Sulfonamides/chemistry , Sulfonamides/pharmacology , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase IX/chemistry , Models, Molecular
2.
Bioconjug Chem ; 35(6): 790-803, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38750635

ABSTRACT

Tumor imaging and delivery of therapeutic agents may be achieved by designing high-affinity and high-selectivity compounds recognizing a tumor cell-expressing biomarker, such as carbonic anhydrase IX (CA IX). The CAIX, overexpressed in many hypoxic solid tumors, helps adjust to the energy requirements of the hypoxic environment, reduces intracellular acidification, and participates in the metastatic invasion of adjacent tissues. Here, we designed a series of sulfonamide compounds bearing CAIX-recognizing, high-affinity, and high-selectivity groups conjugated via a PEG linker to near-infrared (NIR) fluorescent probes used in the clinic for optically guided cancer surgery. We determined compound affinities for CAIX and other 11 catalytically active CA isozymes by the thermal shift assay and showed that the affinity Kd value of CAIX was in the subnanomolar range, hundred to thousand-fold higher than those of other CA isozymes. Similar affinities were also observed for CAIX expressed on the cancer cell surface in live HeLa cell cultures, as determined by the competition assay. The NIR-fluorescent compounds showed excellent properties in visualizing CAIX-positive tumors but not CAIX-negative knockout tumors in a nude mice xenograft model. These compounds would therefore be helpful in optically guided cancer surgery and could potentially be developed for anticancer treatment by radiotherapy.


Subject(s)
Antigens, Neoplasm , Carbonic Anhydrase IX , Carbonic Anhydrase Inhibitors , Fluorescent Dyes , Humans , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase IX/antagonists & inhibitors , Animals , Fluorescent Dyes/chemistry , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase Inhibitors/therapeutic use , Mice , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/analysis , HeLa Cells , Neoplasms/diagnostic imaging , Mice, Nude , Sulfonamides/chemistry , Infrared Rays , Carbonic Anhydrases/metabolism , Optical Imaging/methods
3.
Database (Oxford) ; 20232023 06 08.
Article in English | MEDLINE | ID: mdl-37290059

ABSTRACT

We introduce a protein-ligand binding database (PLBD) that presents thermodynamic and kinetic data of reversible protein interactions with small molecule compounds. The manually curated binding data are linked to protein-ligand crystal structures, enabling structure-thermodynamics correlations to be determined. The database contains over 5500 binding datasets of 556 sulfonamide compound interactions with the 12 catalytically active human carbonic anhydrase isozymes defined by fluorescent thermal shift assay, isothermal titration calorimetry, inhibition of enzymatic activity and surface plasmon resonance. In the PLBD, the intrinsic thermodynamic parameters of interactions are provided, which account for the binding-linked protonation reactions. In addition to the protein-ligand binding affinities, the database provides calorimetrically measured binding enthalpies, providing additional mechanistic understanding. The PLBD can be applied to investigations of protein-ligand recognition and could be integrated into small molecule drug design. Database URL https://plbd.org/.


Subject(s)
Carbonic Anhydrase Inhibitors , Carbonic Anhydrases , Humans , Ligands , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/metabolism , Thermodynamics , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Protein Binding
4.
Sci Rep ; 12(1): 17644, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36271018

ABSTRACT

Numerous human cancers, especially hypoxic solid tumors, express carbonic anhydrase IX (CAIX), a transmembrane protein with its catalytic domain located in the extracellular space. CAIX acidifies the tumor microenvironment, promotes metastases and invasiveness, and is therefore considered a promising anticancer target. We have designed a series of high affinity and high selectivity fluorescein-labeled compounds targeting CAIX to visualize and quantify CAIX expression in cancer cells. The competitive binding model enabled the determination of common CA inhibitors' dissociation constants for CAIX expressed in exponentially growing cancer cells. All tested sulfonamide compounds bound the proliferating cells with similar affinity as to recombinantly purified CAIX. The probes are applicable for the design of selective drug-like compounds for CAIX and the competition strategy could be applied to other drug targets.


Subject(s)
Carbonic Anhydrases , Neoplasms , Humans , Carbonic Anhydrase IX/genetics , Carbonic Anhydrase IX/metabolism , Fluorescent Dyes , Carbonic Anhydrases/metabolism , Cell Line, Tumor , Antigens, Neoplasm/metabolism , Sulfonamides/pharmacology , Fluoresceins
5.
ChemistryOpen ; 10(5): 567-580, 2021 05.
Article in English | MEDLINE | ID: mdl-33945229

ABSTRACT

A key part of the optimization of small molecules in pharmaceutical inhibitor development is to vary the molecular design to enhance complementarity of chemical features of the compound with the positioning of amino acids in the active site of a target enzyme. Typically this involves iterations of synthesis, to modify the compound, and biophysical assay, to assess the outcomes. Selective targeting of the anti-cancer carbonic anhydrase isoform XII (CA XII), this process is challenging because the overall fold is very similar across the twelve CA isoforms. To enhance drug development for CA XII we used a reverse engineering approach where mutation of the key six amino acids in the active site of human CA XII into the CA II isoform was performed to provide a protein chimera (chCA XII) which is amenable to structure-based compound optimization. Through determination of structural detail and affinity measurement of the interaction with over 60 compounds we observed that the compounds that bound CA XII more strongly than CA II, switched their preference and bound more strongly to the engineered chimera, chCA XII, based on CA II, but containing the 6 key amino acids from CA XII, behaved as CA XII in its compound recognition profile. The structures of the compounds in the chimeric active site also resembled those determined for complexes with CA XII, hence validating this protein engineering approach in the development of new inhibitors.


Subject(s)
Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrases/metabolism , Chimera/metabolism , Sulfonamides/chemistry , Amides/chemistry , Amino Acid Sequence , Carbonic Anhydrase Inhibitors/metabolism , Catalytic Domain , Crystallization , Drug Design , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Mutant Proteins , Protein Binding , Protein Conformation , Protein Isoforms , Structure-Activity Relationship , Sulfonamides/pharmacology
6.
Int J Mol Sci ; 23(1)2021 Dec 23.
Article in English | MEDLINE | ID: mdl-35008553

ABSTRACT

Among the twelve catalytically active carbonic anhydrase isozymes present in the human body, the CAIX is highly overexpressed in various solid tumors. The enzyme acidifies the tumor microenvironment enabling invasion and metastatic processes. Therefore, many attempts have been made to design chemical compounds that would exhibit high affinity and selective binding to CAIX over the remaining eleven catalytically active CA isozymes to limit undesired side effects. It has been postulated that such drugs may have anticancer properties and could be used in tumor treatment. Here we have designed a series of compounds, methyl 5-sulfamoyl-benzoates, which bear a primary sulfonamide group, a well-known marker of CA inhibitors, and determined their affinities for all twelve CA isozymes. Variations of substituents on the benzenesulfonamide ring led to compound 4b, which exhibited an extremely high observed binding affinity to CAIX; the Kd was 0.12 nM. The intrinsic dissociation constant, where the binding-linked protonation reactions have been subtracted, reached 0.08 pM. The compound also exhibited more than 100-fold selectivity over the remaining CA isozymes. The X-ray crystallographic structure of compound 3b bound to CAIX showed the structural position, while several structures of compounds bound to other CA isozymes showed structural reasons for compound selectivity towards CAIX. Since this series of compounds possess physicochemical properties suitable for drugs, they may be developed for anticancer therapeutic purposes.


Subject(s)
Benzoates/pharmacology , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase Inhibitors/pharmacology , Sulfonamides/pharmacology , Catalytic Domain/drug effects , Crystallography, X-Ray/methods , Humans , Isoenzymes/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Binding/physiology , Structure-Activity Relationship , Thermodynamics , Tumor Microenvironment/drug effects , Benzenesulfonamides
7.
Eur J Med Chem ; 185: 111825, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31740053

ABSTRACT

By applying an approach of a "ring with two tails", a series of novel inhibitors possessing high-affinity and significant selectivity towards selected carbonic anhydrase (CA) isoforms has been designed. The "ring" consists of 2-chloro/bromo-benzenesulfonamide, where the sulfonamide group is as an anchor coordinating the Zn(II) in the active site of CAs, and halogen atom orients the ring affecting the affinity and selectivity. First "tail" is a substituent containing carbonyl, carboxyl, hydroxyl, ether groups or hydrophilic amide linkage. The second "tail" contains aryl- or alkyl-substituents attached through a sulfanyl or sulfonyl group. Both "tails" are connected to the benzene ring and play a crucial role in selectivity. Varying the substituents, we designed compounds selective for CA VII, CA IX, CA XII, or CA XIV. Since due to binding-linked protonation reactions the binding-ready fractions of the compound and protein are much lower than one, the "intrinsic" affinities were calculated that should be used to study correlations between crystal structures and the thermodynamics of binding for rational drug design. The "intrinsic" affinities together with the intrinsic enthalpies and entropies of binding together with co-crystal structures were used demonstrate structural factors determining major contributions for compound affinity and selectivity.


Subject(s)
Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Sulfonamides/pharmacology , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Halogenation , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Benzenesulfonamides
8.
Eur J Med Chem ; 156: 61-78, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-30006175

ABSTRACT

Rational design of compounds that would bind specific pockets of the target proteins is a difficult task in drug design. The 12 isoforms of catalytically active human carbonic anhydrases (CAs) have highly similar active sites that make it difficult to design inhibitors selective for one or several CA isoforms. A series of CA inhibitors based on 2-chloro/bromo-benzenesulfonamide that is largely fixed in the CA active site together with one or two tails yielded compounds that were synthesized and evaluated as inhibitors of CA isoforms. Introduction of a second tail had significant influence on the binding affinity and two-tailed compounds in most cases provided high affinity and selectivity for CA IX and CA XIV. The contacts between several compounds and CA amino acids were determined by X-ray crystallography. Together with the intrinsic enthalpy and entropy of binding they provided the structure-thermodynamics correlations for this series of compounds with the insight how to rationally build compounds with desired CA isoform as a target.


Subject(s)
Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Drug Design , Sulfonamides/chemistry , Sulfonamides/pharmacology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Binding Sites , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase IX/chemistry , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Catalytic Domain , Crystallography, X-Ray , Halogenation , Humans , Molecular Docking Simulation , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Structure-Activity Relationship , Thermodynamics , Benzenesulfonamides
9.
Bioorg Med Chem ; 26(3): 675-687, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29305297

ABSTRACT

The similarity of human carbonic anhydrase (CA) active sites makes it difficult to design selective inhibitors for one or several CA isoforms that are drug targets. Here we synthesize a series of compounds that are based on 5-[2-(benzimidazol-1-yl)acetyl]-2-chloro-benzenesulfonamide (1a) which demonstrated picomolar binding affinity and significant selectivity for CA isoform five A (VA), and explain the structural influence of inhibitor functional groups to the binding affinity and selectivity. A series of chloro-substituted benzenesulfonamides bearing a heterocyclic tail, together with molecular docking, was used to build inhibitors that explore substituent influence on the binding affinity to the CA VA isoform.


Subject(s)
Benzimidazoles/chemistry , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrases/chemistry , Drug Design , Benzimidazoles/chemical synthesis , Benzimidazoles/metabolism , Binding Sites , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/metabolism , Carbonic Anhydrases/metabolism , Catalytic Domain , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Molecular Docking Simulation , Structure-Activity Relationship
10.
Q Rev Biophys ; 51: e10, 2018 01.
Article in English | MEDLINE | ID: mdl-30912486

ABSTRACT

The aim of rational drug design is to develop small molecules using a quantitative approach to optimize affinity. This should enhance the development of chemical compounds that would specifically, selectively, reversibly, and with high affinity interact with a target protein. It is not yet possible to develop such compounds using computational (i.e., in silico) approach and instead the lead molecules are discovered in high-throughput screening searches of large compound libraries. The main reason why in silico methods are not capable to deliver is our poor understanding of the compound structure-thermodynamics and structure-kinetics correlations. There is a need for databases of intrinsic binding parameters (e.g., the change upon binding in standard Gibbs energy (ΔGint), enthalpy (ΔHint), entropy (ΔSint), volume (ΔVintr), heat capacity (ΔCp,int), association rate (ka,int), and dissociation rate (kd,int)) between a series of closely related proteins and a chemically diverse, but pharmacophoric group-guided library of compounds together with the co-crystal structures that could help explain the structure-energetics correlations and rationally design novel compounds. Assembly of these data will facilitate attempts to provide correlations and train data for modeling of compound binding. Here, we report large datasets of the intrinsic thermodynamic and kinetic data including over 400 primary sulfonamide compound binding to a family of 12 catalytically active human carbonic anhydrases (CA). Thermodynamic parameters have been determined by the fluorescent thermal shift assay, isothermal titration calorimetry, and by the stopped-flow assay of the inhibition of enzymatic activity. Kinetic measurements were performed using surface plasmon resonance. Intrinsic thermodynamic and kinetic parameters of binding were determined by dissecting the binding-linked protonation reactions of the protein and sulfonamide. The compound structure-thermodynamics and kinetics correlations reported here helped to discover compounds that exhibited picomolar affinities, hour-long residence times, and million-fold selectivities over non-target CA isoforms. Drug-lead compounds are suggested for anticancer target CA IX and CA XII, antiglaucoma CA IV, antiobesity CA VA and CA VB, and other isoforms. Together with 85 X-ray crystallographic structures of 60 compounds bound to six CA isoforms, the database should be of help to continue developing the principles of rational target-based drug design.


Subject(s)
Antineoplastic Agents/chemistry , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrases/chemistry , Computer Simulation , Bicarbonates/chemistry , Catalytic Domain , Enzyme Stability , Humans , Kinetics , Molecular Structure , Protein Isoforms/chemistry , Sulfonamides/chemistry , Thermodynamics
11.
ChemMedChem ; 12(2): 161-176, 2017 01 20.
Article in English | MEDLINE | ID: mdl-28001003

ABSTRACT

The goal of rational drug design is to understand structure-thermodynamics correlations in order to predict the chemical structure of a drug that would exhibit excellent affinity and selectivity for a target protein. In this study we explored the contribution of added functionalities of benzenesulfonamide inhibitors to the intrinsic binding affinity, enthalpy, and entropy for recombinant human carbonic anhydrases (CA) CA I, CA II, CA VII, CA IX, CA XII, and CA XIII. The binding enthalpies of compounds possessing similar chemical structures and affinities were found to be very different, spanning a range from -90 to +10 kJ mol-1 , and are compensated by a similar opposing entropy contribution. The intrinsic parameters of binding were determined by subtracting the linked protonation reactions. The sulfonamide group pKa values of the compounds were measured spectrophotometrically, and the protonation enthalpies were measured by isothermal titration calorimetry (ITC). Herein we describe the development of meta- or ortho-substituted fluorinated benzenesulfonamides toward the highly potent compound 10 h, which exhibits an observed dissociation constant value of 43 pm and an intrinsic dissociation constant value of 1.1 pm toward CA IX, an anticancer target that is highly overexpressed in various tumors. Fluorescence thermal shift assays, ITC, and X-ray crystallography were all applied in this work.


Subject(s)
Carbonic Anhydrase Inhibitors/metabolism , Carbonic Anhydrases/metabolism , Sulfonamides/metabolism , Binding Sites , Calorimetry , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrases/chemistry , Catalytic Domain , Crystallography, X-Ray , Halogenation , Humans , Molecular Dynamics Simulation , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry , Thermodynamics , Benzenesulfonamides
12.
J Biotechnol ; 240: 31-42, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27773757

ABSTRACT

Human carbonic anhydrase XIV (CA XIV), a transmembrane protein, highly expressed in the central nervous system, is difficult to recombinantly express and purify in large scale for the measurements of inhibitor binding and drug design. CA XIV belongs to the family of twelve catalytically active CA isoforms in the human body. Disorders in the expression of CA XIV cause serious diseases and CA XIV has been described as a possible drug target for the treatment of epilepsy, some retinopathies, and skin tumors. In this study, the effect of different promoters, E. coli strains, and the length of recombinant CA XIV protein construct were analyzed for the production CA XIV in large scale by using affinity purification. Active site titration by inhibitors and the isothermal titration calorimery revealed over 96% purity of the protein. Enzymatic activity of the purified CA XIV was determined by following the CO2 hydration using the stopped-flow technique. Several inhibitors were discovered that exhibited selectivity towards CA XIV over other CA isoforms and could be developed as drugs.


Subject(s)
Carbonic Anhydrases/biosynthesis , Carbonic Anhydrases/isolation & purification , Escherichia coli , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Escherichia coli/genetics , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
13.
ChemMedChem ; 10(4): 662-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25758852

ABSTRACT

Substituted tri- and tetrafluorobenzenesulfonamides were designed, synthesized, and evaluated as high-affinity and isoform-selective carbonic anhydrase (CA) inhibitors. Their binding affinities for recombinant human CA I, II, VA, VI, VII, XII, and XIII catalytic domains were determined by fluorescent thermal shift assay, isothermal titration calorimetry, and a stopped-flow CO2 hydration assay. Variation of the substituents at the 2-, 3-, and 4-positions yielded compounds with a broad range of binding affinities and isoform selectivities. Several 2,4-substituted-3,5,6-trifluorobenzenesulfonamides were effective CA XIII inhibitors with high selectivity over off-target CA I and CA II. 3,4-Disubstituted-2,5,6-trifluorobenzenesulfonamides bound CAs with higher affinity than 2,4-disubstituted-3,5,6-trifluorobenzenesulfonamides. Many such fluorinated benzenesulfonamides were found to be nanomolar inhibitors of CA II, CA VII, tumor-associated CA IX and CA XII, and CA XIII. X-ray crystal structures of inhibitors bound in the active sites of several CA isoforms provide structure-activity relationship information for inhibitor binding affinities and selectivity.


Subject(s)
Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Sulfonamides/chemistry , Sulfonamides/pharmacology , Carbonic Anhydrases/chemistry , Catalytic Domain , Crystallography, X-Ray , Halogenation , Humans , Models, Molecular , Benzenesulfonamides
14.
J Med Chem ; 57(22): 9435-46, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25358084

ABSTRACT

Human carbonic anhydrase IX (CA IX) is highly expressed in tumor tissues, and its selective inhibition provides a potential target for the treatment of numerous cancers. Development of potent, highly selective inhibitors against this target remains an unmet need in anticancer therapeutics. A series of fluorinated benzenesulfonamides with substituents on the benzene ring was designed and synthesized. Several of these exhibited a highly potent and selective inhibition profile against CA IX. Three fluorine atoms significantly increased the affinity by withdrawing electrons and lowering the pKa of the benzenesulfonamide group. The bulky ortho substituents, such as cyclooctyl or even cyclododecyl groups, fit into the hydrophobic pocket in the active site of CA IX but not CA II, as shown by the compound's co-crystal structure with chimeric CA IX. The strongest inhibitor of recombinant human CA IX's catalytic domain in human cells achieved an affinity of 50 pM. However, the high affinity diminished the selectivity. The most selective compound for CA IX exhibited 10 nM affinity. The compound that showed the best balance between affinity and selectivity bound with 1 nM affinity. The inhibitors described in this work provide the basis for novel anticancer therapeutics targeting CA IX.


Subject(s)
Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrases/chemistry , Drug Design , Benzene/chemistry , Calorimetry , Carbon Dioxide/chemistry , Carbonic Anhydrase IV/chemistry , Catalysis , Catalytic Domain , Crystallization , Crystallography, X-Ray , Humans , Hydrogen-Ion Concentration , Kinetics , Neoplasms/drug therapy , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Sulfonamides/chemistry , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL