WO2017206041A1 - Method for determining endurance of interaction between ligand and target protein in cell - Google Patents

Method for determining endurance of interaction between ligand and target protein in cell Download PDF

Info

Publication number
WO2017206041A1
WO2017206041A1 PCT/CN2016/083998 CN2016083998W WO2017206041A1 WO 2017206041 A1 WO2017206041 A1 WO 2017206041A1 CN 2016083998 W CN2016083998 W CN 2016083998W WO 2017206041 A1 WO2017206041 A1 WO 2017206041A1
Authority
WO
WIPO (PCT)
Prior art keywords
ligand
target protein
cell
intracellular
dissociation
Prior art date
Application number
PCT/CN2016/083998
Other languages
French (fr)
Chinese (zh)
Inventor
王帅
Original Assignee
王帅
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 王帅 filed Critical 王帅
Priority to CN201680003155.8A priority Critical patent/CN109690316A/en
Priority to PCT/CN2016/083998 priority patent/WO2017206041A1/en
Publication of WO2017206041A1 publication Critical patent/WO2017206041A1/en

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids

Definitions

  • the invention belongs to the technical field of protein binding, and in particular relates to a method for determining the endurance of interaction of a target protein in a cell.
  • the residence time of the drug target protein in vitro is not equal to the retention time of the drug target protein in the cell or in vivo, and it is also uncertain whether the long-lasting drug target protein retention time in vitro can be converted into a sustained pharmacological action in the cell or in vivo, especially The long-term efficacy of quantification.
  • the retention time of the drug target protein in the cell is longer than the retention time of the drug target protein in vitro.
  • the retention time of the drug target protein in the cell is a new parameter different from the retention time of the drug target protein in vitro, and is used to describe the persistence of the drug target protein interaction at the cell level.
  • it is still difficult to measure the retention time of drug target proteins in the body. Therefore, it is more reasonable to predict the persistence of the drug in vivo by using the intracellular drug target protein retention time measured under physiological conditions.
  • the covalently bound fluorescently labeled probe molecule has the same binding site as the ligand to be detected and can be used in a competitive manner to study the persistence of interaction of the intracellular ligand with the target protein.
  • U.S. Patent Application Serial No. 14/104,860 the disclosure of which is incorporated herein by reference to the entire disclosure of the disclosure of the disclosure of the disclosure of the disclosure of the disclosure of the disclosure of the disclosure of the disclosure of the disclosure of The persistence of the interaction of the body with the target protein.
  • the above two methods for applying probe molecules have the following problems: 1) it is very difficult or even impossible to design and synthesize a suitable fluorescently labeled probe molecule for each target protein; 2) it takes time for the probe molecule to enter the cell to reach equilibrium. About half an hour, so the above two methods can not accurately record the initial stage of the dissociation reaction; 3) especially because the fluorescent probe molecules can interfere with the interaction of the ligand with the target protein through competitive inhibition or allosteric regulation. . Therefore, the above two methods cannot accurately measure the endurance of intracellular ligand target protein interactions.
  • Protein thermal shift assays have been widely used in academia and industry to evaluate the interaction of ligand molecules with purified target proteins. Moreau et al., Mol. Biosyst., 6, 1285-1292, 2010, report a green fluorescent protein-based thermal transfer method that can be used to study ligands in cell lysates using non-purified GFP-fused target proteins. Interaction of GFP-fused target proteins.
  • PCT/GB2012/050853 discloses a method for determining intracellular ligand binding to a target protein using a thermal transfer technique, which can be used to study natural unpurified target proteins in cells, including: 1) Whether the body can interact with intracellular target proteins, 2) the affinity of intracellular ligand target protein interactions.
  • a method for determining the endurance of a ligand protein interaction in a cell comprising the steps of:
  • Step 1 treating the cells with the ligand
  • Step 2 removing the ligand outside the extracellular
  • Step 3 collecting the cells at different time points after removing the ligand
  • Step 4 heating the cells
  • Step 5 lysing the cells
  • step 6 a function of the level of soluble or insoluble target protein in the cell lysate as a function of time is analyzed.
  • the ligand is added to the cell culture medium and enters the cell to interact with the target protein.
  • the extracellular ligand is removed by replacing the fresh medium to break the balance between the extracellular and intracellular ligands, thereby triggering the dissociation reaction of the intracellular ligand.
  • the cells at different time points after removal of extracellular ligands were collected, and the level of ligand-binding target protein in cells at different time points after removal of extracellular ligands was determined by cell thermal transfer technique to construct the dissociation of intracellular ligand target protein interactions.
  • the curve calculates the dissociation rate constant, retention time and dissociation half-life of the intracellular ligand target protein interaction, and quantitatively describes the endurance of the intracellular ligand target protein interaction.
  • the method of the present invention determines the kinetic profile of the intracellular ligand target protein occupancy, calculates the therapeutic duration of the ligand, and explains the persistence of the pharmacological effects of the intracellular ligand from the perspective of binding persistence.
  • the present invention relates to the binding endurance of intracellular ligand target protein interactions, from the angle of time The degree of interaction of intracellular ligands with target proteins was investigated.
  • the method of the present invention can be used to screen compounds, and find suitable binding endactive active molecules as seed molecules, which can be used to guide the optimization of lead compounds, and to design and synthesize drugs with desired binding endurance.
  • the methods of the invention can be used to measure and compare the binding potency of a drug to a plurality of intracellular target proteins, such as target proteins and off-target proteins, wild-type and mutants, to determine the selectivity of the intracellular target protein of the drug from a time perspective.
  • the method of the present invention can be used to measure and compare the binding persistence of drug and target protein interactions in different types of cells, such as different types of tumor cells, different tissue cells, etc., and determine the cell selectivity of the drug from a time perspective. .
  • the methods of the present invention can be used to predict and design the most effective dosing regimen, particularly by setting a suitable dosing interval to reduce off-target related side effects caused by drug exposure depending on the duration of treatment of the drug.
  • the ligand is a cell metabolite, a small molecule or a drug
  • the target protein is a protein molecule that interacts with a ligand in the cell
  • the cell is a mammalian cell, a cell strain, an engineered cell or a primary cell.
  • the term "ligand” as used in this specification refers to a molecule or compound to be detected that is capable of interacting with a target protein in a cell.
  • the ligand can be a polypeptide, a nucleic acid, a substrate for an enzyme, a cellular metabolite or a hormone.
  • the ligand is preferably a pharmacologically active compound, a seed molecule, a lead compound, a drug candidate or a drug.
  • the ligand may also be a prodrug that binds to a target protein after being converted into a drug by hydrolysis or an enzymatic reaction or the like.
  • the ligand may be naturally occurring or unnaturally chemically synthesized.
  • the ligand described in this specification is not limited to its size or structure.
  • target protein refers to a protein molecule that interacts with a ligand in a cell.
  • the target protein can be any protein of the cell, including proteins of the cell membrane, cytoplasm, nucleus or other subcellular organelles.
  • the interaction of the ligand with the target protein can occur within the cell, for example, the ligand interacts with the intracellular kinase; interaction of the ligand with the target protein can occur on the cell membrane, for example, the ligand interacts with the extramembranous portion of the membrane protein.
  • the target protein may be naturally occurring or may be recombinantly expressed after transfection of a plasmid or vector encoding the target protein.
  • the target protein can be expressed as a fusion protein with a tag molecule or polypeptide, such as a FLAG tag, a His tag, a GFP tag, a GST tag or a luciferase.
  • a tag molecule or polypeptide such as a FLAG tag, a His tag, a GFP tag, a GST tag or a luciferase.
  • Target proteins within cells include GPCR receptors, ion channels, proteases, kinases, nuclear receptors, transcription factors or enzymes.
  • Target egg White can be wild type or mutant.
  • the target protein in the present invention is not limited to its type or kind.
  • cell refers to a cell or similar structure containing a target protein that interacts with a ligand.
  • Cells include prokaryotic cells such as bacteria, eukaryotic cells such as yeast, single cell eukaryotes such as Leishmania, insect cells, mammalian cells, engineered cells or cell lines. Purified subcellular organs such as mitochondria can also function as cells.
  • the cells are isolated from cultured primary cells of the animal or human, and more preferably cultured under physiological or pathological conditions close to the body.
  • Ligand target protein interactions may have different binding endurance in different cells due to different cellular environments, so the method of the invention determines the cell selectivity of the ligand from a time perspective.
  • the method for determining the endurance of the ligand interaction of the ligand in the cell wherein the amount of the ligand added in the step 1 is determined according to one of the following:
  • Mode 1 determined according to the EC50 value of the effect of the ligand on the cells, wherein the concentration of the ligand in the medium in the step 1 is 0.5-20 times the EC50 value;
  • the concentration of the ligand in the medium in the step 1 is 70%-100% of the ligand in the cell to the target protein occupancy;
  • the concentration of the ligand in the medium in the step 1 is the maximum blood concentration and the minimum effective concentration of the ligand in the animal or human body. Range concentration range.
  • the treatment time of the ligand to the cells in step 1 is determined according to the binding rate of the ligand to the intracellular target protein, and the treatment of the ligand is terminated when the intracellular ligand target protein interaction reaches equilibrium.
  • the ligand In the first step of the present invention, after the ligand is added to the cell culture medium, the ligand enters the cell by diffusion, active or passive transport, or endocytosis, disperses into different subcellular organelles, and finally interacts with the target protein to establish Ligand target protein complex.
  • the level or amount of the intracellular ligand target protein complex is closely related to the concentration of the ligand (hereinafter referred to as the test concentration of the ligand) and the action time added to the medium.
  • the test concentration of the ligand may be in any concentration range as long as sufficient ligand target protein complex can be produced.
  • the test concentration of the ligand can be determined according to its cellular activity, and can produce at least 5%, 10%, 20%, 30%, 50%, 70%, 80%, 90% or 100% cell activity, Or the test concentration of the ligand is equal to at least 0.5, 1, 2, 4, 5, 10, 20 or more EC50 values.
  • the test concentration of the ligand is preferably determined according to its intracellular target protein occupancy rate, and can produce at least 5%, 10%, 20%, 30%, 50%, 70%, 80%, 90% or 100% of the target protein. Occupancy, and more preferably at least 70%, 80%, 90% or 100% target protein occupancy.
  • the test concentration of the ligand is preferably determined based on its metabolic kinetics in the animal or human, such as the maximum plasma concentration, or the lowest effective concentration in the therapeutic concentration range.
  • the action time of the ligand is determined by the rate of binding of the ligand to the target protein in the cell, and the treatment of the ligand is terminated when the intracellular ligand target protein interaction reaches equilibrium. For example, it can be 10, 30, 60 minutes, 2, 4, 6, 12 hours or longer, usually 30 minutes to 1 hour.
  • cell viability should be monitored throughout the dissociation test, especially for high activity compounds at high test concentrations and long dissociation times, which may result in cell death, correspondingly Adjust the dose of the ligand and the time of the dissociation reaction.
  • the ligand molecule staurosporin at a test concentration of 10 ⁇ M can cause significant apoptosis 6 hours after removal of extracellular staurosporine.
  • the method for determining the endurance of the ligand interaction of the ligand in the cell is: replacing the fresh medium or washing the cells with the solution and then replacing the fresh medium.
  • the removal of the ligand in the cell culture medium in step 2 of the present invention is to break the balance between the intracellular and extracellular ligands, thereby triggering the dissociation reaction of the intracellular ligand target protein.
  • To remove the extracellular ligand it is preferred to replace the fresh cell culture medium, and it is better to wash the cells with the solution for 1, 2 or 3 times before replacing the fresh medium.
  • other methods include the addition of chemicals that specifically react or bind to the ligand, or dilution with a large amount of fresh medium.
  • the intracellular ligand Upon removal of the extracellular ligand, the intracellular ligand will leave the cell and diffuse into the culture medium, referred to as the remaining ligand.
  • the target protein occupancy of the remaining ligand In order to initiate an intracellular dissociation reaction, the target protein occupancy of the remaining ligand must be significantly less than the target protein occupancy of the starting ligand, and preferably the target protein occupancy of the remaining ligand is very small and negligible.
  • the remaining ligand In the case of a high activity ligand at high test concentrations, for example 10,000 times the equilibrium dissociation constant value (Kd), the remaining ligand may result in a false positive strong binding endurance test result. In this case, a more thorough extracellular ligand clearance should be used. Order, for example 3, 4, 5 or more cleanings.
  • the method for determining the endurance of the ligand interaction of the ligand in the cell wherein the time point of collecting the cells in the step 3 is at least 8 time points, and the interval between adjacent time points is based on the solution of the ligand target protein complex The rate of departure is determined.
  • step 3 of the present invention cells at different time points after removal of the extracellular ligand are collected, representing different stages of the dissociation reaction of the intracellular ligand target protein.
  • the number of time points of the dissociation reaction may be, but not limited to, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 20 or more, and preferably includes at least 8 data points.
  • the time interval between different time points is determined according to the dissociation rate of the ligand target protein complex, which may be 1, 2, 3, 5, 10, 20, 30, 60 minutes; or 2, 3, 4, 6 , 12, 24, 48 hours or more.
  • the methods of the invention also include collecting cells prior to removal of the extracellular ligand, representing the maximum initial level of ligand target protein interaction in the dissociation reaction.
  • the intracellular ligand target protein complex will be rapidly dissociated after removal of the extracellular ligand, in which case more data should be set in the early stages of the dissociation reaction. Point, especially the analysis of cells before removal of extracellular ligands.
  • the ligand target protein complex will slowly disintegrate after removal of the extracellular ligand, in which case sufficient data points should be set to cover the entire dissociation at appropriate intervals. reaction process.
  • the methods of the invention may further comprise a negative control and a positive control.
  • the positive control is preferably a cell treated with a ligand but not having a ligand removed, having a known level of cell activity or a known target protein occupancy, and more preferably treated with the same test concentration of the ligand but Unremoved cells represent the maximum level of ligand-bound target protein under this assay condition.
  • the negative control reflects the level of noise under this test condition, which may be cells without ligand treatment, and is preferably cells treated with the remaining ligands described above.
  • the negative control may also be a cell that is sufficiently long after removal of the extracellular ligand until the intracellular ligand target protein complex is completely dissociated.
  • the method for determining the endurance of the interaction of the ligand protein in the cell wherein the temperature at which the cells are heat treated in step 4 is higher than the initial melting temperature of the ligand not bound to the target protein and And below the final melting temperature of the ligand binding target protein.
  • the level of the intracellular ligand-bound target protein at different time points after removal of the extracellular ligand is detected by a cell thermal transfer technique.
  • the heating temperature may be a temperature above the initial melting temperature of the unbound target protein of the ligand but below the final melting temperature interval of the ligand-bound target protein.
  • the heating temperature is preferably the temperature at which the soluble target protein has the most significant difference in the negative control and sample.
  • the heating time can be any time range, such as 0.5, 1, 2, 3 or 5 minutes, as long as the level of soluble target protein in the negative control and the sample is significantly different in this case, and it is better to have the largest The difference in degree.
  • Heating instruments include PCR machines, incubators, water baths, etc., as long as the cells can be heated to a specific temperature.
  • the method of cell lysis in the step 5 of the present invention may be any method capable of lysing cells to release intracellular proteins.
  • the cell lysis method may be a sonication method, preferably a freeze-thaw method comprising 1, 2 or 3 cycles, and more preferably a cell lysate containing an ionic or non-ionic denaturing agent or amphiphilic molecule.
  • Cells are lysed, especially membrane proteins, such as 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% NP-40.
  • the cell lysate further contains a protease inhibitor, an enzyme cofactor such as Mg 2+ , or a redox agent such as DTT for stabilization and Protect the target protein.
  • soluble and insoluble protein components can be separated by some separation technique.
  • the protein separation technique can be any method capable of separating soluble and insoluble protein components.
  • the protein separation technique is preferably centrifugal separation, the supernatant is a soluble protein component, and the precipitate is an insoluble protein component. Filtration separation is also preferred, the filtrate is a soluble protein component and the residue is an insoluble protein component.
  • the soluble target protein can also be isolated by affinity purification of the target protein-specific antibody.
  • the method for determining the endurance of the interaction of the ligand protein in the cell comprises antibody technology, mass spectrometry, biological activity of the target protein or enzymatic activity of the fusion tag.
  • the soluble or insoluble protein component is analyzed to determine the amount of target protein in which it is soluble or insoluble.
  • To measure an insoluble target protein it is necessary to first dissolve the insoluble protein component. It is therefore preferred to measure soluble target proteins.
  • Quantitative methods for target proteins include mass spectrometry, such as orbitrap or ion traps; application of target protein or fusion tag-specific antibody methods, such as immunoblotting, ELISA; and quantitative analysis using the biological activity of the target protein or the enzymatic activity of the fusion tag.
  • the best method for quantifying target proteins is to use high-throughput methods for directly detecting the level of soluble target proteins in cell lysates, such as alpha scanning, fluorescence resonance energy transfer, and fluorescence polarization.
  • the step 6 further comprises: analyzing the soluble target protein, determining the level of the target protein bound by the ligand in the cell, and constructing the cell as a function of the dissociation time.
  • the dissociation curve of the internal ligand target protein interaction calculates the dissociation rate constant, residence time and dissociation half-life of the ligand ligand protein interaction in the cell.
  • the soluble protein component is analyzed, the target protein soluble therein is measured, the amount of the target protein bound by the intracellular ligand is determined, and the dissociation curve of the intracellular ligand target protein interaction is constructed.
  • a higher level of soluble target protein was detected in the sample cells than in the negative control representing the noise level, meaning that the ligand still binds to the intracellular target protein at this time point after removal of the extracellular ligand.
  • the level of ligand-bound target protein is equal to the difference between the sample and the soluble target protein in the negative control, initially reflecting the strength of the endurance of the intracellular ligand target protein interaction.
  • the ligand-unbound target protein at some heating temperatures can be completely unfolded and precipitated.
  • the level of soluble target protein directly reflects the intracellular ligand binding.
  • the level of the target protein The dissociation curve of intracellular ligand target protein interactions can be constructed as a function of the dissociation time as a function of the dissociation time by removing the amount of target protein bound by the intracellular ligand at different time points after removal of the extracellular ligand.
  • the dissociation rate and retention time of intracellular ligand target protein interactions can be determined based on the dissociation curve of the intracellular ligand target protein, quantitatively describing the endurance of the intracellular ligand target protein interaction.
  • the dissociation reaction of the intracellular ligand target protein may also include a plurality of dissociation phases having significantly different dissociation rates, so that it is difficult to accurately apply the conventional dissociation rate and residence time in this case.
  • the persistence of intracellular ligand target protein interactions was compared (Examples 1 and 2).
  • the method of the present invention advances One step comprises determining the time required for dissociation of 50% of the ligand target protein complex in the cell according to the dissociation curve of the intracellular ligand target protein, which is called the dissociation half-life of the intracellular ligand target protein, and can be used to describe intracellular distribution.
  • the persistence of body target protein interactions Similarly, the time required for dissociation of different percentages of ligand target protein complexes can also be determined, for example 10%, 20%, 30%, 40%, for more precise description of the persistence of intracellular ligand target protein interactions. force.
  • the step 6 further comprises determining the target protein occupancy rate at different time points after removing the extracellular ligand, and constructing the kinetics of the ligand protein occupancy rate in the cell.
  • the curve calculates the duration of treatment of the ligand above the lowest effective target protein occupancy.
  • Ligand has strong binding endurance with intracellular target proteins, meaning that the ligand has the potential to produce long-lasting intracellular pharmacological effects, however in some cases, for example, different doses of administration, even the same ligand target protein binding Persistence can still lead to different intracellular pharmacological effects over time.
  • the method of the invention further comprises determining the occupancy of the ligand target protein and the level of the target protein bound by the intracellular ligand in the positive control.
  • the lowest effective target protein occupancy that produces intracellular pharmacology can be determined based on the biological properties of the target protein and the pathogenic mechanism.
  • the time range in which the intracellular ligand target protein occupancy kinetic curve is higher than the lowest effective ligand target protein occupancy rate is the time range in which the intracellular pharmacological action can be sustained after the extracellular ligand is eliminated.
  • the invention is referred to as the "Therapeutic duration time range" of the ligand.
  • Therapeutic duration time range of the ligand.
  • there may be different kinetic curves of the intracellular ligand target protein occupancy and there may be different durations of intracellular pharmacological action of the ligand.
  • the target protein occupancy rate of the dissociation reaction is 100%
  • the dissociation curve of the intracellular ligand target protein is directly equal to the kinetic curve of the intracellular ligand target protein occupancy rate, in which case the treatment of the ligand
  • the duration range can also be determined from the dissociation curve of the intracellular ligand target protein.
  • the method for determining the persistence of interaction of a ligand protein in a cell, and the dissociation reaction of an intracellular ligand target protein interaction comprises dissociation reaction during removal of the extracellular ligand and removal of the extracellular ligand Dissociation reaction.
  • the method of the present invention it is found that the dissociation reaction of the intracellular ligand target protein can be apparently occurred in the process of removing the extracellular ligand, in which case it is possible to ignore the dissociation reaction during removal of the extracellular ligand, possibly This leads to erroneous conclusions about the strength of the binding endurance of the ligand target protein in the cell.
  • the method of the invention further comprises analyzing the cells prior to removal of the extracellular ligand as a positive control representing the ligand-bound target protein in the dissociation assay.
  • the maximum initial level determines the extent to which the intracellular ligand target protein complex dissociates during removal of the extracellular ligand. As shown in Example 1, if the cells before removal of the extracellular ligand are not analyzed and the dissociation reaction during removal of the extracellular ligand is neglected, the second stage of the dissociation reaction may be mistaken for the entire intracellular ligand target. The dissociation reaction of the protein results in a strong binding endurance of the false positive.
  • intracellular target proteins are not completely homogeneous components, and actually contain target proteins of different conformations, different post-translationally modified target proteins, different subcellular organelle target proteins, and Target proteins that interact with different binding partners may have different fast or slow dissociation rates for target proteins and ligand molecules in different states.
  • intracellular ligand-bound or unbound target proteins may be degraded, new unbound target proteins may be synthesized, and degradation and synthesis of intracellular target proteins may attenuate intracellular distribution.
  • the binding endurance of the body target protein Similar to cell proliferation can also lead to reduction The persistence of ligand-protein interactions in weak cells.
  • Dissociation assays due to intracellular ligand target proteins may persist for a long period of time, especially for ligands with slow dissociation rates, such as 48 or 96 hours.
  • the cells continue to grow during the dissociation reaction, so in order to accurately compare the levels of intracellular ligand-bound target proteins at different time points, the method of the present invention further includes normalization.
  • the standard for normalization can be based on the protein concentration of the soluble cell lysate or on the number of cells in the sample. Proliferation of cells reduces the average level of ligand-target protein complexes in cells, which in turn leads to a decrease in the persistence of ligand-protein interactions in cells.
  • the rate of cell proliferation in vivo is usually significantly lower than the rate of cell proliferation in vitro. In this case, the survival of the ligand target protein interaction is underestimated due to the rapid cell proliferation rate in vitro, so in order to evaluate the endurance of the ligand interaction of the intracellular ligand in the case of proximity to the body, it is necessary to exclude the in vitro
  • cells of the same sample volume such as 1 ml or 1 well, can be used as normalization criteria. In this case, samples at different time points may contain different numbers of cells but possess the same initial level of ligand target protein complex. To some extent, it is also understood that the cell proliferation rate is 0 in this case. Example 3).
  • This normalization standard is especially suitable for cells with slow proliferation rate in vivo, which can avoid the insufficiency of ligand cell target interaction due to in vitro cell proliferation, and better reflect the endurance of ligand target protein interaction in vivo.
  • the method of the invention can be used for the development of new drugs, determining the binding endurance of intracellular target proteins of different compounds, selecting suitable binding endurance compounds as seed molecules, lead compounds, drug candidates according to the biological characteristics and pathological characteristics of the target protein.
  • drugs Strongly potent compounds have higher levels of soluble target protein at the same dissociation time point than weakly durable compounds, as well as slower intracellular dissociation rates and longer intracellular residence times. In particular, it has a longer duration of treatment.
  • the binding endurance parameter of the compound can be used to guide the optimization of the lead compound and to design and synthesize a drug molecule having the desired binding endurance.
  • the persistence of intracellular compound target protein interactions can be used as a new parameter to further sort and distinguish compounds from a time perspective, which is helpful for selection. The most active molecule.
  • the method of the present invention can be used to compare the binding endurance of a compound to a different target protein in a cell, and to evaluate the selectivity of a target protein in a compound cell from a time perspective, which is called the selectivity of a target protein in time.
  • Active molecules usually interact with multiple proteins in the cell, of which the target protein is responsible for pharmacological activity, and the other is called off-target protein.
  • the binding of active molecules to off-target proteins can sometimes lead to deleterious side effects, so the target protein selectivity of the drug is an important goal in the optimization phase of the lead compound.
  • the traditional target protein selectivity is mainly to compare the affinity of the active molecule to the target protein and the off-target protein from a thermodynamic point of view.
  • the intracellular target protein selectivity of the present invention refers to the kinetics of target protein occupancy and off-target protein occupancy in the cell, comparing target protein occupancy and off-target protein at the same time point.
  • the occupancy rate, or the retention time of the target protein in the cell and the retention time of the off-target protein determine the intracellular target protein selectivity of the active molecule from the kinetic point of view.
  • the target protein selectivity of the active molecule can be further evaluated from the kinetic point of view using the intracellular target protein of the present invention.
  • the methods of the invention can also be used to compare the binding endurance of an active molecule to a wild-type and mutant target protein, determining the selectivity of the active molecule for wild-type and mutant from a time perspective.
  • the method of the present invention can be used to compare the persistence of an active compound interacting with a target protein in different cells, and to evaluate the cell selectivity of the active molecule from a time perspective.
  • the target protein can be distributed in multiple tissues and organs in the body, such as heart, lung, kidney, liver, brain, etc., wherein the tissue related to the pharmacological activity of the active molecule is called a target tissue, and the active molecule interacts with a target protein in other tissues. May cause undesirable side effects.
  • target proteins in different tissues have the same amino acid sequence, different cellular environments may result in the persistence of interactions between different active molecular target proteins.
  • the present invention evaluates the selectivity of active molecules in different tissue cells from a time perspective to help explain the mechanism of action of the drug, design a rational dosing regimen, and reduce the side effects associated with off-target tissue.
  • the method of the present invention can be used to compare the persistence of active compounds interacting with target proteins in different tumor cells, and to evaluate the selectivity of tumor cells of active molecules from a time perspective, and to help predict drugs in different tumors. The efficacy of the design of personalized dosing regimens.
  • the methods of the present invention can be used to predict the most effective dosing regimen for design in an animal or human, particularly in predicting the time interval of administration based on the duration of treatment of the drug.
  • Dosing regimens including dosing and dosing intervals, have a significant impact on the efficacy and toxicity of bioactive molecules in animals and humans.
  • the time interval for administration is usually determined based on the pharmacokinetics of the compound, and it is considered that once the blood concentration of the drug is below the therapeutic concentration range, the pharmacological activity of the drug will be immediately terminated.
  • the present inventors have demonstrated that certain compounds can interact with intracellular target proteins at about 50% target protein occupancy even after removal of extracellular compounds (Examples 2 & 3), meaning that the cellular activity of the compound is even if the cells are removed.
  • the compound is still present 24 hours after the compound. It is more scientific and rational to predict the time interval of administration in combination with the pharmacological kinetic properties of the drug in combination with the therapeutic duration of the drug of the present invention, and thus the method of the present invention facilitates the design of an optimal dosing regimen,
  • the rate of cell dissociation maintains a sustained target protein occupancy and achieves therapeutic efficacy, and the toxic side effects associated with the off-target protein are attenuated by appropriate dosing intervals, thereby obtaining the largest therapeutic window of the drug.
  • a compound can be completely eliminated within 2 hours in the body, however in this case there is a 24 hour treatment duration range.
  • the compound requires multiple administrations per day, however, considering the therapeutic duration of the compound, the once-daily dosing regimen can be reduced.
  • the method of the present invention does not require the use of fluorescent probe molecules, so the method of the present invention can accurately measure the endurance of intracellular ligand target protein interaction without the interference effect brought by the probe molecule; the method of the present invention has no probe molecule The delayed effect can completely record the dissociation reaction of the ligand target protein in the cell.
  • the method of the invention does not require the construction of a fusion protein, which can detect native proteins, especially directly from the patient.
  • the method of the invention is a versatile method for the analysis of the vast majority of ligands and target proteins. Using this approach, it was found that the intracellular ligand target protein dissociation reaction is much more complex than the reported data and can contain multiple dissociation phases with significantly different dissociation rates. Most importantly, the intracellular ligand target protein occupancy was maintained at about 60% 24 hours after removal of the extracellular ligand.
  • the method of the invention represents an effective method for determining the persistence of interactions of target protein proteins within a cell.
  • Figure 1 is a graph of the thermal melting of Aurora A in Jurkat cells in the absence (•) and presence ( ⁇ ) staurosporine.
  • Figure 2 is a graph showing the dose response of Aurora A and staurosporine in Jurkat cells.
  • Figure 3 is a dissociation plot of the interaction of staurosporin Aurora A in Jurkat cells, normalized to the same number of cells at each time point.
  • Figure 4 is a kinetic plot of the occupancy of staurosporin Aurora A in Jurkat cells, normalized to the same number of cells at each time point.
  • Figure 5 is a graph of the thermal melting of PARP-1 in Jurkat cells in the absence (•) and presence ( ⁇ ) Talazoparib.
  • Figure 6 is a graph showing the dose response of Talazoparib and PARP-1 in Jurkat cells.
  • Figure 7 is a dissociation plot of Talazoparib interaction with PARP-1 in Jurkat cells, normalized to the same number of cells at each time point.
  • Figure 8 is a graph showing the kinetics of Talazoparib and PARP-1 occupancy in Jurka cells, the normalization criterion being the same number of cells at each time point.
  • Figure 9 is a dissociation plot of Talazoparib interaction with PARP-1 in Jurkat cells, normalized to the same volume of cells at each time point, simulating a cell proliferation rate of zero.
  • Figure 10 is a kinetic curve of Talazoparib and PARP-1 occupancy in Jurkat cells.
  • the normalization criterion is the same volume of cells at each time point, simulating a cell growth rate of zero.
  • Jurkat cells were cultured in a RPMI 1640 medium at 37 ° C in a 5% carbon dioxide incubator.
  • Jurkat cells were treated with 10 ⁇ M staurosporin (#19-123MG, Merck KGaA, Germany) for half an hour in serum-free RPMI1640 medium, and Jurkat cells treated with 0.1% DMSO in parallel were used as negative controls.
  • 1x10 6 cells were collected from each data point and resuspended in 20 ⁇ L of PBS, heated by PCR instrument (Eppendorf, Germany) at different temperatures in the temperature range of 37-64 ° C for three minutes, and then containing protease inhibitors (Roche, Switzerland)
  • the cell lysate 1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5
  • the cell lysate 1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5
  • Aurora A was detected by SDS-PAGE and immunoblotting.
  • the thermal melting curve of Aurora A in Jurkat cells under conditions of no treatment with staurosporine was constructed relative to the level of Aurora A at the lowest heating temperature.
  • Dose-effect assay Jurkat cells were treated with different concentrations of staurosporine from 0.1 nM to 10 ⁇ M in serum-free RPMI 1640 medium for 1 hour; DMSO concentration was 0.1%. 1 ⁇ 10 6 cells were resuspended in 20 ⁇ L of PBS at each concentration point, and heated by a PCR machine at 58 ° C for three minutes. The cells were then lysed on ice for half an hour with a cell lysate containing protease inhibitor (Roche, Switzerland) (1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5). After centrifugation, the supernatant was collected and Aurora A was detected by SDS-PAGE and immunoblotting. The level of Aurora A in the negative control was subtracted from the sample as the level of staurosporine-conjugated Aurora A, and a dose-response curve of intracellular staurosporine and Aurora A was constructed.
  • Dissociation of intracellular staurosporin Aurora A interaction by time as a function of the level of Aurora A in the negative control from the sample as the level of staurosporine-conjugated Aurora A in the sample The curve determines the dissociation rate, retention time and dissociation half-life of intracellular staurosporin Aurora A interaction. Further, based on the known occupancy rate of staurosporin Aurora A in the positive control, the kinetic curve of intracellular staurosporin Aurora A occupancy in this test was determined, and the staurosporin was determined under the test conditions. Duration of treatment.
  • FIG. 1 is a graph of the thermal melting of Aurora A in Jurkat cells in the absence (•) and presence ( ⁇ ) staurosporin.
  • ( ⁇ ) indicates a thermal melting curve of Aurora A in the case of no treatment with staurosporine
  • ( ⁇ ) indicates a thermal melting curve of Aurora A in the case of treatment with staurosporine.
  • the ordinate "relative strip intensity” refers to the ratio of the Aurora A luminescence intensity to the strongest Aurora A luminescence intensity of each data result measured by immunoblotting in Example 1. According to the Aurora A thermal melting curve shown in Fig.
  • the staurosporin treatment group exhibited a higher level of Aurora A than the control group at a temperature interval of 55-61 ° C, so that 58 ° C can be selected as the heating temperature for the cells.
  • Dissociation test of endosporin and Aurora A Dissociation test of endosporin and Aurora A.
  • FIG 2 is a graph of the dose response of intracellular Aurora A and staurosporine.
  • the level of intracellular staurosporin-conjugated Aurora A increases with the concentration of staurosporine (in the range of 0.1-10000 nM), so a concentration of 100 nM staurosporine can be selected.
  • the concentration of staurosporine in the range of 0.1-10000 nM
  • Figure 3 is a dissociation curve of the interaction of staurosporin Aurora A in Jurkat cells
  • Figure 4 is a kinetic plot of the occupancy of staurosporin Aurora A in Jurkat cells.
  • the normalization criterion is the same number of cells at each time point.
  • the level of staurosporin-conjugated Aurora A decreased non-linearly with increasing dissociation time.
  • a two-phase dissociation model was used to analyze the dissociation curve of staurosporin Aurora A in Jurkat cells (shown in Figure 3).
  • the first phase is a very fast dissociation phase that occurs during the removal of extracellular staurosporine, comparing cell samples prior to removal of extracellular staurosporine and removal of extracellular staurosporin A dissociation time point sample found that about 70% of the staurosporin Aurora A complex dissociated during about 5 minutes, and it was difficult to accurately fit the phase solution due to the very fast dissociation rate and limited data points. Rate constant.
  • the second phase is a slow dissociation phase, and approximately 26% of the staurosporin Aurora A complex gradually dissociates in the next 36 hours.
  • the dissociation rate constant of this phase is 0.042 h -1 , presumably Jurkat
  • the retention time of intracellular staurosporin Aurora A interaction is about 36 hours, or longer, but its dissociation half-life is only about 0.1 hour.
  • the intracellular staurosporin dissociation curve (shown in Figure 3) can be converted to the occupancy of staurosporin Aurora A in Jurkat cells under the test conditions.
  • a kinetic curve (shown in Figure 4) from which the time range over which a certain Aurora A occupancy is exceeded under the test conditions can be determined.
  • the purpose of this experiment was to determine the persistence of the Talazoparib PARP-1 interaction in Jurkat cells and found that intracellular Talazoparib has a strong binding endurance with PARP-1.
  • Jurkat cells were cultured in a RPMI 1640 medium at 37 ° C in a 5% carbon dioxide incubator.
  • Jurkat cells were treated with 1 ⁇ M Talazoparib (Selleckchem, USA) in serum-free RPMI 1640 medium for half an hour, and cells treated with 0.1% DMSO in parallel served as a negative control.
  • 1x10 6 cells were collected from each data point and resuspended in 20 ⁇ L of PBS, heated with a PCR instrument (Eppendorf, Germany) at different temperatures ranging from 37-55 ° C for three minutes, and then containing a protease inhibitor (Roche, Switzerland).
  • Dose-effect test Jurkat cells were treated with Talazoparib at different concentrations from 0.001 nM to 10 ⁇ M in serum-free RPMI 1640 medium for 0.5 hour at a DMSO concentration of 0.1%. 1 ⁇ 10 6 cells were collected from each concentration point and resuspended in 20 ⁇ L of PBS, and heated at 49 ° C for three minutes using a PCR machine. The cells were then lysed on ice for half an hour with a cell lysate containing protease inhibitor (Roche, Switzerland) (1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5).
  • protease inhibitor Roche, Switzerland
  • the level of PARP-1 in the sample was subtracted from the level of PARP-1 in the negative control as the level of Talazoparib-bound PARP-1 in the sample, and the dissociation curve of the Talazoparib PARP-1 interaction in Jurkat cells was constructed as a function of time to determine the cell. Dissociation rate, retention time and dissociation half-life of the internal Talazoparib PARP-1 interaction. Based on the known Talazoparib PARP-1 occupancy rate in the positive control, the dissociation curve of intracellular Talazoparib PARP-1 interaction was converted to the kinetic curve of intracellular Talazoparib PARP-1 occupancy under the test conditions, and the experimental conditions were determined. The duration of treatment of the lower Talazoparib.
  • SDS-PAG & Immunoblotting Separation of PARP-1 protein by SDS-PAGE Trans-Blot Turbo (Bio-Rad, USA) was transferred onto a PVDF membrane, blocked, and tested with anti-PARP-1 antibody (#sc-8007, SANTA CRUZ BIOTECHNOLOGY, USA) with a CCD camera (GE Healthcare, USA) Record the chemiluminescent signal.
  • FIG. 5 is a graph of the thermal melting of PARP-1 in Jurkat cells in the absence (•) and in the presence of ( ⁇ ) Talazoparib; wherein ( ⁇ ) indicates the heat of PARP-1 in the absence of treatment with Talazoparib Melt curve; ( ⁇ ) shows the thermal melting curve of PARP-1 in the case of Talazoparib treatment; wherein, the ordinate "relative band intensity” refers to the PARP-1 luminescence intensity of each data result measured by immunoblotting in Example 2. The ratio of the intensity of the strongest PARP-1 luminescence.
  • the Talazoparib treatment group exhibited a higher level of PARP-1 than the control group at a temperature range of 46-49 ° C, so 48.5 or 49 ° C could be selected as the heating temperature for intracellular determination.
  • FIG. 6 is a graph of the dose response of Talazoparib and PARP-1 in Jurkat cells.
  • the level of soluble PARP-1 and Talazoparib (in the range of 0.001-10000 nM) showed a concentration-dependent increase, so 100 nM Talazoparib was selected, corresponding to 93% PARP-1 occupancy in cells, as dissociation Test concentration of Talazoparib in the test.
  • Figure 7 is a dissociation plot of the interaction of Talazoparib with PARP-1 in Jurkat cells.
  • Figure 8 is a graph showing the kinetics of Talazoparib and PARP-1 occupancy in Jurka cells.
  • the normalization criteria of Figures 7 and 8 are the same number of cells at each time point.
  • the level of Talazoparib-bound PARP-1 decreased non-linearly with increasing dissociation time.
  • a three-phase dissociation model was used to analyze the dissociation curve of Talazoparib interaction with PARP-1 in Jurkat cells (as shown in Figure 7).
  • the first phase is a fast dissociation phase that occurs during the first hour after removal of extracellular Talazoparib, during which about 50% of the Talazoparib PARP-1 complex dissociates with a dissociation rate constant of 0.69 h -1 . And during the removal of extracellular Talazoparib, no dissociation of intracellular Talazoparib PARP-1 complex was observed.
  • the second phase is a very slow dissociation phase that occurs during the next 23 hours during which there is little change in the level of intracellular Talazoparib PARP-1 complex, and its dissociation rate is very slow and difficult to fit accurately.
  • the third phase is a slightly faster dissociation phase, and the remaining Talazoparib PARP-1 complex in the cell will gradually dissociate.
  • the intracellular dissociation curve of Talazoparib (shown in Figure 7) was converted to the kinetic curve of Talazoparib PARP-1 occupancy in Jurkat cells under the experimental conditions (Fig. 8).
  • the duration of treatment of Talazoparib under the test conditions can be determined from the PARP-1 occupancy kinetics curve. For example, assuming that the lowest effective PARP-1 occupancy in the cell is 50%, in this case the treatment duration of Talazoparib is in the range of 24 hours, meaning that the cell activity of Talazoparib can continue to be maintained for 24 hours after the removal of extracellular Talazoparib. .
  • this example was to demonstrate that cell proliferation can attenuate the persistence of intracellular ligand target protein interactions and the duration of intracellular pharmacological effects.
  • this experiment uses a fixed cell sampling volume as a normalization standard to simulate a cell proliferation rate of 0.
  • the sample may contain a different number of cells at each time point, but Ligand target protein complexes with the same starting level.
  • Jurkat cells were treated with 50 nM Talazoparib serum-free RPMI 1640 medium for 0.5 hour with a DMSO concentration of 0.1%. Extracellular Talazoparib was removed by replacing fresh RPMI1640 medium containing 10% serum. Then, 1 ml of cells were collected at each detection time point, counted, resuspended in 20 ⁇ L of PBS, and heated by a PCR instrument at 48.5 ° C for three minutes. Cell lysate (1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5) containing protease inhibitor (Roche, Switzerland) was lysed on ice for half an hour.
  • the dissociation curve of the Talazoparib PARP-1 interaction in the Jurkat cells determines the dissociation rate, retention time and dissociation half-life of the intracellular Talazoparib PARP-1 interaction. Based on the known Talazoparib PARP-1 occupancy rate in the positive control, the dissociation curve of intracellular Talazoparib PARP-1 interaction was converted to the kinetic curve of intracellular Talazoparib PARP-1 occupancy under the test conditions, and the experimental conditions were determined. The duration of treatment of the lower Talazoparib.
  • the PARP-1 protein was separated by SDS-PAGE and transferred to a PVDF membrane using Trans-Blot Turbo (Bio-Rad, USA). After blocking, the chemiluminescent signal was recorded with a CCD camera (GE Healthcare, USA) using an anti-PARP-1 antibody assay (#sc-8007, SANTA CRUZ BIOTECHNOLOGY, USA).
  • Figure 9 is a dissociation plot of Talazoparib interaction with PARP-1 in Jurkat cells.
  • the normalization criterion is the same volume of cells at each time point, simulating a cell growth rate of zero.
  • Figure 10 is a graph showing the kinetics of Talazoparib and PARP-1 occupancy in Jurkat cells.
  • the normalization standard is the same volume of cells at each time point, and the simulated cell growth rate is zero.
  • the level of Talazoparib-bound PARP-1 decreases non-linearly with increasing dissociation time.
  • a dissociation curve of the interaction between Talazoparib and PARP-1 in Jurkat cells under the experimental conditions of this example was analyzed using a two-phase dissociation model.
  • the first phase is a fast dissociation phase that occurs during the first hour after removal of extracellular Talazoparib, during which about 50% of the Talazoparib PARP-1 complex dissociates with a dissociation rate constant of 0.62 h -1 . It was confirmed again that no dissociation of the intracellular Talazoparib PARP-1 complex was observed during the removal of extracellular Talazoparib.
  • the second phase is a very slow dissociation phase that occurs during the next 35 hours during which there is little change in the level of intracellular Talazoparib PARP-1 complex, and the dissociation rate is very slow and difficult to fit accurately.
  • about 60% of the Talazoparib PARP-1 complex was still present at the 36-hour time point, so the retention time of Talazoparib interacting with PARP-1 in Jurkat cells was much longer than 36 hours under this test condition.
  • Intracellular Talazoparib and PARP The -1 dissociation half-life is also more than 36 hours.
  • the intracellular dissociation curve of Talazoparib (shown in Figure 9) was converted to the kinetic curve of Talazoparib PARP-1 occupancy in Jurkat cells under the experimental conditions (Fig. 10).
  • the duration of treatment of Talazoparib in this case can be determined from the PARP-1 occupancy kinetic curve.
  • the least potent PARP-1 occupancy rate in the cell is 50%, so the duration of Talazoparib treatment is more than 36 hours under this test condition, meaning that the cell viability of Talazoparib can continue to be maintained after removal of extracellular Talazoparib. hour.
  • Example 2 uses the normalization method of the same cell number to represent the normal cell proliferation rate
  • Example 3 uses the normalization method of the fixed cell sampling volume to simulate the case where the cell proliferation rate is zero. Comparing the dissociation curves of Talazoparib PARP-1 in both cases (as shown in Figures 7 and 9), especially at the 36 hour time point, and the dissociation half-life, Example 3 was found to be stronger than Example 2.
  • the persistence of intracellular Talazoparib PARP-1 interactions demonstrates that cell proliferation can attenuate the persistence of ligand-target protein interactions in cells.
  • Example 3 Comparing the kinetic curves of Talazoparib PARP-1 occupancy (as shown in Figures 8 and 10) and the duration of treatment in both cases, it can be found that Example 3 is still more effective than Example 2 even if a slightly lower Talazoparib test concentration is applied. With a longer duration of treatment, it is demonstrated that cell proliferation can attenuate the duration of cellular pharmacological effects.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Analytical Chemistry (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

Disclosed is a method for determining the endurance of the interaction between a ligand and a target protein in a cell, comprising: (1) treating the cell with the ligand; (2) removing the ligand outside the cell; (3) collecting the cell at different time points after the ligand is removed; (4) heating the cell; (5) lysing the cell; and (6) analysing the level of soluble or insoluble target protein in a cell lysis solution as a function of time. The method can be used to determine the dissociation rate constant, residence time and dissociation half-life of the interaction between the ligand and the target protein in the cell and the time range of therapeutic duration of the ligand so as to quantitatively describe the endurance of the interaction between the ligand and the target protein in the cell and to predict the duration of pharmacological action in the cell. The method can be used to develop new drugs and discover drugs with desired binding endurance. The method can be used to design a dosage regimen, especially a time interval of drug delivery.

Description

测定细胞内配体靶蛋白相互作用持久力的方法Method for determining the endurance of ligand interactions in intracellular ligands 技术领域Technical field
本发明属于蛋白结合技术领域,具体涉及一种测定细胞内配体靶蛋白相互作用持久力的方法。The invention belongs to the technical field of protein binding, and in particular relates to a method for determining the endurance of interaction of a target protein in a cell.
背景技术Background technique
有些药物即使已从体内循环系统中消除很长时间,仍然具有令人惊奇的持久的疗效。然而现在仍然不知道什么样的药物能够拥有如此持久的疗效以及如何设计此类药物。为了解释这个矛盾的药理学现象,Robert A.Copeland等人在Nat.Rev.Drug Discov.,5,730-739,2006中提出了药物靶蛋白滞留时间模型,认为药物的疗效主要与药物靶蛋白相互作用持续时间的长短有关。目前药物靶蛋白滞留时间主要是用一些生物物理和生物化学的实验技术测量,例如表面等离子共振和荧光偏振等,主要与药物靶蛋白相互作用的解离速率相关。然而体外的药物靶蛋白滞留时间并不等于细胞内的或体内的药物靶蛋白滞留时间,并且也不确定体外持久的药物靶蛋白滞留时间能否转换成细胞内或体内持久的药理作用,尤其是定量的多长时间的持久疗效。为了解决这个问题,首先需要能够准确的测量细胞内或体内的药物靶蛋白滞留时间,尤其是确定细胞内或体内药物靶蛋白占有率的动力学。Some drugs have surprisingly long-lasting effects even if they have been eliminated from the circulatory system for a long time. However, it is still not known what kind of drugs can have such long-lasting effects and how to design such drugs. To explain this contradictory pharmacological phenomenon, Robert A. Copeland et al., in Nat. Rev. Drug Discov., 5, 730-739, 2006, proposed a drug target protein retention time model, suggesting that the drug's efficacy mainly interacts with drug target proteins. The duration is related to the length of time. At present, drug target protein retention time is mainly measured by some biophysical and biochemical experimental techniques, such as surface plasmon resonance and fluorescence polarization, which are mainly related to the dissociation rate of drug target protein interaction. However, the residence time of the drug target protein in vitro is not equal to the retention time of the drug target protein in the cell or in vivo, and it is also uncertain whether the long-lasting drug target protein retention time in vitro can be converted into a sustained pharmacological action in the cell or in vivo, especially The long-term efficacy of quantification. In order to solve this problem, it is first necessary to accurately measure the retention time of drug target proteins in cells or in vivo, especially to determine the kinetics of drug target protein occupancy in cells or in vivo.
由于药物与靶蛋白的相互作用通常发生在细胞的表面或细胞的内部,细胞内的生理环境比体外的实验条件要复杂的多,所以细胞内药物靶蛋白滞留时间比体外的药物靶蛋白滞留时间会受到更多因素的影响,包括1)药物靶蛋白相互作用的解离速率,2)不同构象的靶蛋白,3)药物的再结合,4)与其它蛋白结合的药物,5)靶蛋白的天然底物,翻译后修饰以及结合伙伴,6)药物的主动转运,7)药物与靶蛋白的亚细胞分布,8)细胞增殖,9)靶蛋白的降解与合成。由此可见,细胞内药物靶蛋白滞留时间是不同于体外的药物靶蛋白滞留时间的一个新的参数,用于描述细胞层面上的药物靶蛋白相互作用的持久力。由于技术上的原因,目前仍很难测量体内的药物靶蛋白滞留时间, 所以应用生理条件下测量的细胞内药物靶蛋白滞留时间预测药物体内疗效的持久性会具有更高的合理性。然而目前仍然缺少可准确评价细胞内药物靶蛋白相互作用的持久力,确定细胞内药物靶蛋白滞留时间的方法。Since the interaction between the drug and the target protein usually occurs on the surface of the cell or inside the cell, the physiological environment inside the cell is much more complicated than the experimental conditions in vitro, so the retention time of the drug target protein in the cell is longer than the retention time of the drug target protein in vitro. Will be affected by more factors, including 1) the rate of dissociation of drug target protein interactions, 2) target proteins of different conformations, 3) recombination of drugs, 4) drugs that bind to other proteins, and 5) target proteins Natural substrates, post-translational modifications and binding partners, 6) active transport of drugs, 7) subcellular distribution of drugs and target proteins, 8) cell proliferation, 9) degradation and synthesis of target proteins. It can be seen that the retention time of the drug target protein in the cell is a new parameter different from the retention time of the drug target protein in vitro, and is used to describe the persistence of the drug target protein interaction at the cell level. For technical reasons, it is still difficult to measure the retention time of drug target proteins in the body. Therefore, it is more reasonable to predict the persistence of the drug in vivo by using the intracellular drug target protein retention time measured under physiological conditions. However, there is still a lack of methods for accurately assessing the persistence of intracellular drug target protein interactions and determining the retention time of intracellular drug target proteins.
对于已报道的评价细胞内药物靶蛋白的结合持久力的方法,每一个方法有自己的优点和缺点。传统上主要是应用放射性同位素标记的药物分子测量其细胞内滞留时间(Expert Opin.Drug Discov.,7,583-595,2012),该方法虽然简单但费用极高,而且实验操作复杂,更重要的是该方法不能区分所检测到的放射性信号是来自与靶蛋白结合的还是与脱靶蛋白结合的药物分子。Each method has its own advantages and disadvantages for the reported methods of assessing the binding endurance of drug target proteins in cells. Traditionally, radioisotope-labeled drug molecules have been used to measure their intracellular residence time (Expert Opin. Drug Discov., 7, 583-595, 2012). This method is simple but extremely expensive, and the experimental operation is complicated. More importantly, This method does not distinguish whether the detected radioactive signal is from a drug molecule that binds to the target protein or binds to the off-target protein.
共价结合荧光标记的探针分子与待检测的配体有相同的结合位点,可通过竞争的方式用于研究细胞内配体与靶蛋白相互作用的持久力。Bradshaw等人在Nat.Chem.Biol.,11,525-531,2015中报道应用此种方法研究BTK抑制剂细胞内结合持久力的特性。另外,申请号14/104,860的美国专利公开了一种用荧光素酶融合的靶蛋白和荧光标记的探针分子的生物发光共振能量转移(BRET)的方法,也是通过竞争的方式检测细胞内配体与靶蛋白相互作用的持久力。上述两种应用探针分子的方法有以下问题:1)非常困难甚至不可能为每一个靶蛋白设计合成一个合适的荧光标记的探针分子;2)由于探针分子进入细胞达到平衡状态需要花费约半个小时时间,因此以上两种方法不能准确的记录解离反应的初始阶段;3)尤其是由于荧光探针分子可通过竞争抑制或变构调节的方式干扰配体与靶蛋白的相互作用。因此以上两种方法不能准确的测量细胞内配体靶蛋白相互作用的持久力。The covalently bound fluorescently labeled probe molecule has the same binding site as the ligand to be detected and can be used in a competitive manner to study the persistence of interaction of the intracellular ligand with the target protein. Bradshaw et al., Nat. Chem. Biol., 11, 525-531, 2015, report the use of this method to study the intracellular binding endurance properties of BTK inhibitors. In addition, U.S. Patent Application Serial No. 14/104,860, the disclosure of which is incorporated herein by reference to the entire disclosure of the disclosure of the disclosure of the disclosure of the disclosure of the disclosure of the disclosure of the disclosure of The persistence of the interaction of the body with the target protein. The above two methods for applying probe molecules have the following problems: 1) it is very difficult or even impossible to design and synthesize a suitable fluorescently labeled probe molecule for each target protein; 2) it takes time for the probe molecule to enter the cell to reach equilibrium. About half an hour, so the above two methods can not accurately record the initial stage of the dissociation reaction; 3) especially because the fluorescent probe molecules can interfere with the interaction of the ligand with the target protein through competitive inhibition or allosteric regulation. . Therefore, the above two methods cannot accurately measure the endurance of intracellular ligand target protein interactions.
蛋白热转移技术(thermal shift assay)已经广泛的应用于学术界和工业界评价配体分子和纯化的靶蛋白的相互作用。Moreau等人在Mol.Biosyst.,6,1285-1292,2010中报道了一种基于绿色荧光蛋白的热转移方法,该方法可用非纯化的GFP融合的靶蛋白,研究细胞裂解液中配体和GFP融合的靶蛋白的相互作用。此外,PCT/GB2012/050853公开了一种使用热转移技术测定细胞内配体与靶蛋白结合的方法(cellular thermal shift assay),可研究细胞内天然的未纯化的靶蛋白,包括:1)配体能否与细胞内靶蛋白相互作用,2)细胞内配体靶蛋白相互作用的亲和力。然而,以上提及的所有的热转移试验都是 在平衡的试验条件下进行的,即整个试验过程中配体和靶蛋白的浓度都保持不变。此外,以上提及的所有的热转移试验测量都需要在一个短的时间内完成,如推荐的半个小时,以防止靶蛋白的降解或合成(Nat.Protoc.,9,2100-2122,2014)。Protein thermal shift assays have been widely used in academia and industry to evaluate the interaction of ligand molecules with purified target proteins. Moreau et al., Mol. Biosyst., 6, 1285-1292, 2010, report a green fluorescent protein-based thermal transfer method that can be used to study ligands in cell lysates using non-purified GFP-fused target proteins. Interaction of GFP-fused target proteins. In addition, PCT/GB2012/050853 discloses a method for determining intracellular ligand binding to a target protein using a thermal transfer technique, which can be used to study natural unpurified target proteins in cells, including: 1) Whether the body can interact with intracellular target proteins, 2) the affinity of intracellular ligand target protein interactions. However, all the heat transfer tests mentioned above are The concentration of ligand and target protein remained unchanged under the equilibrium test conditions, ie throughout the test. In addition, all of the heat transfer test measurements mentioned above need to be completed in a short period of time, such as the recommended half hour, to prevent degradation or synthesis of the target protein (Nat. Protoc., 9, 2100-2122, 2014). ).
发明内容Summary of the invention
本发明的目的是,提供一种测定细胞内配体靶蛋白相互作用持久力的方法。It is an object of the present invention to provide a method for determining the persistence of interactions of ligands in a cell with a target protein.
本发明为实现上述目的所采用的技术方案如下:The technical solution adopted by the present invention to achieve the above object is as follows:
一种测定细胞内配体靶蛋白相互作用持久力的方法,该方法包括如下步骤:A method for determining the endurance of a ligand protein interaction in a cell, the method comprising the steps of:
步骤1,用所述配体处理所述细胞; Step 1, treating the cells with the ligand;
步骤2,去除所述细胞外的所述配体;Step 2, removing the ligand outside the extracellular;
步骤3,收集去除所述配体后不同时间点的所述细胞;Step 3, collecting the cells at different time points after removing the ligand;
步骤4,加热所述细胞;Step 4, heating the cells;
步骤5,裂解所述细胞; Step 5, lysing the cells;
步骤6,分析细胞裂解液中可溶的或不可溶的所述靶蛋白的水平随时间变化的函数。In step 6, a function of the level of soluble or insoluble target protein in the cell lysate as a function of time is analyzed.
根据本发明的方法,将配体加入到细胞培养基中,进入细胞后与靶蛋白相互作用。通过更换新鲜培养基去除细胞外配体,打破细胞外和细胞内配体的平衡,以此引发细胞内配体的解离反应。收集去除细胞外配体后不同时间点的细胞,应用细胞热转移技术确定去除细胞外配体后不同时间点细胞内配体结合靶蛋白的水平,构建细胞内配体靶蛋白相互作用的解离曲线,计算细胞内配体靶蛋白相互作用的解离速率常数,滞留时间和解离半衰期,定量的描述细胞内配体靶蛋白相互作用的持久力。本发明的方法尤其是可确定细胞内配体靶蛋白占有率的动力学曲线,计算配体的治疗持续时间范围,从结合持久力的角度解释细胞内配体的药理作用的持久性。According to the method of the present invention, the ligand is added to the cell culture medium and enters the cell to interact with the target protein. The extracellular ligand is removed by replacing the fresh medium to break the balance between the extracellular and intracellular ligands, thereby triggering the dissociation reaction of the intracellular ligand. The cells at different time points after removal of extracellular ligands were collected, and the level of ligand-binding target protein in cells at different time points after removal of extracellular ligands was determined by cell thermal transfer technique to construct the dissociation of intracellular ligand target protein interactions. The curve calculates the dissociation rate constant, retention time and dissociation half-life of the intracellular ligand target protein interaction, and quantitatively describes the endurance of the intracellular ligand target protein interaction. In particular, the method of the present invention determines the kinetic profile of the intracellular ligand target protein occupancy, calculates the therapeutic duration of the ligand, and explains the persistence of the pharmacological effects of the intracellular ligand from the perspective of binding persistence.
本发明涉及细胞内配体靶蛋白相互作用的结合持久力,从时间的角 度研究细胞内配体与靶蛋白相互作用的强度。本发明的方法可用于筛选化合物,发现合适的结合持久力的活性分子作为苗头分子,可用于指导先导化合物优化,设计合成具有期望的结合持久力的药物。本发明的方法可用于测量和比较药物与多个细胞内靶蛋白的结合持久力,例如靶蛋白和脱靶蛋白,野生型和突变体,从时间的角度确定药物的细胞内靶蛋白的选择性。本发明的方法可用于测量和比较在不同类型的细胞中,例如不同类型的肿瘤细胞,不同的组织细胞等,药物和靶蛋白相互作用的结合持久力,从时间的角度确定药物的细胞选择性。本发明的方法可用于预测和设计最有效的给药方案,尤其是依据药物的治疗持续时间范围设置合适的给药间隔降低药物暴露量导致的脱靶相关的副作用。The present invention relates to the binding endurance of intracellular ligand target protein interactions, from the angle of time The degree of interaction of intracellular ligands with target proteins was investigated. The method of the present invention can be used to screen compounds, and find suitable binding endactive active molecules as seed molecules, which can be used to guide the optimization of lead compounds, and to design and synthesize drugs with desired binding endurance. The methods of the invention can be used to measure and compare the binding potency of a drug to a plurality of intracellular target proteins, such as target proteins and off-target proteins, wild-type and mutants, to determine the selectivity of the intracellular target protein of the drug from a time perspective. The method of the present invention can be used to measure and compare the binding persistence of drug and target protein interactions in different types of cells, such as different types of tumor cells, different tissue cells, etc., and determine the cell selectivity of the drug from a time perspective. . The methods of the present invention can be used to predict and design the most effective dosing regimen, particularly by setting a suitable dosing interval to reduce off-target related side effects caused by drug exposure depending on the duration of treatment of the drug.
优选地,所述配体是细胞代谢物,小分子或药物;所述靶蛋白是细胞内与配体相互作用的蛋白分子;所述细胞是哺乳动物细胞,细胞株,工程细胞或原代细胞。Preferably, the ligand is a cell metabolite, a small molecule or a drug; the target protein is a protein molecule that interacts with a ligand in the cell; the cell is a mammalian cell, a cell strain, an engineered cell or a primary cell. .
在本说明书中所用的“配体”一词是指一种能够与细胞内靶蛋白相互作用的待检测分子或化合物。配体可以是多肽,核酸,酶的底物,细胞代谢物或荷尔蒙。配体较好的是药理活性的化合物,苗头分子,先导化合物,候选药物或药物。配体也可以是通过水解或酶反应等在细胞内转换为药物后与靶蛋白结合的前药。配体可以是自然出现的或非自然化学合成的。本说明书所描述的配体并不局限于它的大小或结构。The term "ligand" as used in this specification refers to a molecule or compound to be detected that is capable of interacting with a target protein in a cell. The ligand can be a polypeptide, a nucleic acid, a substrate for an enzyme, a cellular metabolite or a hormone. The ligand is preferably a pharmacologically active compound, a seed molecule, a lead compound, a drug candidate or a drug. The ligand may also be a prodrug that binds to a target protein after being converted into a drug by hydrolysis or an enzymatic reaction or the like. The ligand may be naturally occurring or unnaturally chemically synthesized. The ligand described in this specification is not limited to its size or structure.
在本说明书中所用的“靶蛋白”一词是指一种细胞内能与配体相互作用的蛋白分子。靶蛋白可以是细胞的任何一种蛋白,包括细胞膜,细胞质,细胞核或者其它亚细胞器的蛋白。配体与靶蛋白相互作用可发生在细胞内部,例如配体与细胞内激酶相互作用;配体与靶蛋白相互作用可发生在细胞膜上,例如配体与细胞膜蛋白的膜外部分相互作用。靶蛋白可以是自然出现的,也可以是转染编码靶蛋白的质粒或载体后重组表达的。靶蛋白可以与标签分子或多肽作为融合蛋白表达,例如FLAG标签,His标签,GFP标签,GST标签或荧光素酶。细胞内的靶蛋白包括GPCR受体,离子通道,蛋白酶,激酶,核受体,转录因子或酶。靶蛋 白可以是野生型或突变体。本发明中的靶蛋白不局限于其类型或种类。The term "target protein" as used in this specification refers to a protein molecule that interacts with a ligand in a cell. The target protein can be any protein of the cell, including proteins of the cell membrane, cytoplasm, nucleus or other subcellular organelles. The interaction of the ligand with the target protein can occur within the cell, for example, the ligand interacts with the intracellular kinase; interaction of the ligand with the target protein can occur on the cell membrane, for example, the ligand interacts with the extramembranous portion of the membrane protein. The target protein may be naturally occurring or may be recombinantly expressed after transfection of a plasmid or vector encoding the target protein. The target protein can be expressed as a fusion protein with a tag molecule or polypeptide, such as a FLAG tag, a His tag, a GFP tag, a GST tag or a luciferase. Target proteins within cells include GPCR receptors, ion channels, proteases, kinases, nuclear receptors, transcription factors or enzymes. Target egg White can be wild type or mutant. The target protein in the present invention is not limited to its type or kind.
在本说明书中所用的“细胞”一词是指一种含有与配体相互作用的靶蛋白的细胞或类似的结构。细胞包括原核细胞例如细菌,真核细胞例如酵母,单细胞真核生物例如利什曼原虫,昆虫细胞,哺乳动物细胞,工程细胞或细胞株。纯化的亚细胞器例如线粒体,也可以起到类似细胞的作用。细胞较好的是从动物或人分离培养的原代细胞,而更好的是在接近体内生理或病理条件下培养。配体靶蛋白相互作用在不同的细胞中由于不同的细胞环境可有不同的结合持久力,所以本发明的方法可从时间的角度确定配体的细胞选择性。The term "cell" as used in this specification refers to a cell or similar structure containing a target protein that interacts with a ligand. Cells include prokaryotic cells such as bacteria, eukaryotic cells such as yeast, single cell eukaryotes such as Leishmania, insect cells, mammalian cells, engineered cells or cell lines. Purified subcellular organs such as mitochondria can also function as cells. Preferably, the cells are isolated from cultured primary cells of the animal or human, and more preferably cultured under physiological or pathological conditions close to the body. Ligand target protein interactions may have different binding endurance in different cells due to different cellular environments, so the method of the invention determines the cell selectivity of the ligand from a time perspective.
优选地,测定细胞内配体靶蛋白相互作用持久力的方法,其步骤1中配体的加入量根据以下其中之一的方式确定:Preferably, the method for determining the endurance of the ligand interaction of the ligand in the cell, wherein the amount of the ligand added in the step 1 is determined according to one of the following:
方式1,根据配体对细胞产生效应的EC50值确定,所述步骤1中配体在培养基中的浓度为所述EC50值的0.5-20倍; Mode 1, determined according to the EC50 value of the effect of the ligand on the cells, wherein the concentration of the ligand in the medium in the step 1 is 0.5-20 times the EC50 value;
方式2,根据配体在细胞内对靶蛋白占有率确定,所述步骤1中配体在培养基中的浓度为所述配体在细胞内对靶蛋白占有率的70%-100%;2, according to the ligand to the target protein occupancy rate in the cell, the concentration of the ligand in the medium in the step 1 is 70%-100% of the ligand in the cell to the target protein occupancy;
方式3,根据配体在动物或人体内的代谢动力学确定,所述步骤1中配体在培养基中的浓度为所述配体在动物或人体内的最大血药浓度和最低有效浓度的区间浓度范围。Mode 3, according to the metabolic kinetics of the ligand in the animal or human body, the concentration of the ligand in the medium in the step 1 is the maximum blood concentration and the minimum effective concentration of the ligand in the animal or human body. Range concentration range.
优选地,步骤1中配体对细胞的处理时间根据配体与细胞内靶蛋白的结合速率决定,当细胞内配体靶蛋白相互作用达到平衡时结束配体对细胞的处理。Preferably, the treatment time of the ligand to the cells in step 1 is determined according to the binding rate of the ligand to the intracellular target protein, and the treatment of the ligand is terminated when the intracellular ligand target protein interaction reaches equilibrium.
本发明步骤1中,将配体加入到细胞培养基后,配体通过扩散,主动或被动转运,或细胞内吞等方式进入细胞,分散到不同的亚细胞器,最后与靶蛋白相互作用,建立配体靶蛋白复合体。细胞内配体靶蛋白复合体的水平或量与加入到培养基中的配体浓度(以下称配体的试验浓度)和作用时间密切相关。配体的试验浓度可以是任意浓度范围,只要可以产生足够的配体靶蛋白复合体。配体的试验浓度可根据其细胞活性确定,可产生至少5%,10%,20%,30%,50%,70%,80%,90%或100%细胞活性, 或配体的试验浓度等于至少0.5,1,2,4,5,10,20或者更多倍EC50值。配体的试验浓度较好的是根据其细胞内靶蛋白占有率确定,可产生至少5%,10%,20%,30%,50%,70%,80%,90%或100%靶蛋白占有率,而更好的是至少70%,80%,90%或100%靶蛋白占有率。配体的试验浓度最好的是根据其在动物或人体内的代谢动力学确定,例如最大的血药浓度,或治疗浓度范围中的最低有效浓度。配体的作用时间根据配体与细胞内靶蛋白的结合速率决定,当细胞内配体靶蛋白相互作用达到平衡时结束配体对细胞的处理。例如可以是10,30,60分钟,2,4,6,12小时或更长时间,一般是30分钟到1个小时。此外,整个解离试验的过程中应该监测细胞的活力,尤其是对于一些高活性的化合物在高的试验浓度和长的解离时间的条件下,此种情况下可能导致细胞死亡,应相应地调整配体剂量和解离反应的时间。例如本发明实施例1中配体分子星形孢菌素在10μM试验浓度的情况下,在去除细胞外星形孢菌素后6小时可导致明显的细胞凋亡。In the first step of the present invention, after the ligand is added to the cell culture medium, the ligand enters the cell by diffusion, active or passive transport, or endocytosis, disperses into different subcellular organelles, and finally interacts with the target protein to establish Ligand target protein complex. The level or amount of the intracellular ligand target protein complex is closely related to the concentration of the ligand (hereinafter referred to as the test concentration of the ligand) and the action time added to the medium. The test concentration of the ligand may be in any concentration range as long as sufficient ligand target protein complex can be produced. The test concentration of the ligand can be determined according to its cellular activity, and can produce at least 5%, 10%, 20%, 30%, 50%, 70%, 80%, 90% or 100% cell activity, Or the test concentration of the ligand is equal to at least 0.5, 1, 2, 4, 5, 10, 20 or more EC50 values. The test concentration of the ligand is preferably determined according to its intracellular target protein occupancy rate, and can produce at least 5%, 10%, 20%, 30%, 50%, 70%, 80%, 90% or 100% of the target protein. Occupancy, and more preferably at least 70%, 80%, 90% or 100% target protein occupancy. The test concentration of the ligand is preferably determined based on its metabolic kinetics in the animal or human, such as the maximum plasma concentration, or the lowest effective concentration in the therapeutic concentration range. The action time of the ligand is determined by the rate of binding of the ligand to the target protein in the cell, and the treatment of the ligand is terminated when the intracellular ligand target protein interaction reaches equilibrium. For example, it can be 10, 30, 60 minutes, 2, 4, 6, 12 hours or longer, usually 30 minutes to 1 hour. In addition, cell viability should be monitored throughout the dissociation test, especially for high activity compounds at high test concentrations and long dissociation times, which may result in cell death, correspondingly Adjust the dose of the ligand and the time of the dissociation reaction. For example, in the case of the present invention, the ligand molecule staurosporin at a test concentration of 10 μM can cause significant apoptosis 6 hours after removal of extracellular staurosporine.
优选地,测定细胞内配体靶蛋白相互作用持久力的方法,其步骤2中去除配体的方法为:更换新鲜的培养基或者用溶液清洗细胞后再更换新鲜培养基。Preferably, the method for determining the endurance of the ligand interaction of the ligand in the cell, the method of removing the ligand in the step 2 is: replacing the fresh medium or washing the cells with the solution and then replacing the fresh medium.
本发明步骤2中去除细胞培养基中的配体是为了打破细胞内和细胞外配体的平衡,以此引发细胞内配体靶蛋白的解离反应。去除细胞外配体的方法,较好的是更换新鲜的细胞培养基,而更好的是用溶液清洗细胞1,2或3次后再更换新鲜培养基。此外,其它的方法包括添加可与配体特异反应或结合的化学物质,或者用大量的新鲜培养基进行稀释处理。清除细胞外配体后,细胞内配体将离开细胞,扩散到培养基中,称为剩余的配体。为了引发细胞内解离反应,剩余配体的靶蛋白占有率必须显著少于起始配体的靶蛋白占有率,最好的是剩余配体的靶蛋白占有率非常小可以忽略不计。一个高活性的配体在高的试验浓度的情况下,例如10000倍平衡解离常数值(Kd),剩余配体可能会导致假阳性的强结合持久力的试验结果。在此种情况下,应使用更加彻底的细胞外配体清除程 序,例如3,4,5或更多次的清洗。The removal of the ligand in the cell culture medium in step 2 of the present invention is to break the balance between the intracellular and extracellular ligands, thereby triggering the dissociation reaction of the intracellular ligand target protein. To remove the extracellular ligand, it is preferred to replace the fresh cell culture medium, and it is better to wash the cells with the solution for 1, 2 or 3 times before replacing the fresh medium. In addition, other methods include the addition of chemicals that specifically react or bind to the ligand, or dilution with a large amount of fresh medium. Upon removal of the extracellular ligand, the intracellular ligand will leave the cell and diffuse into the culture medium, referred to as the remaining ligand. In order to initiate an intracellular dissociation reaction, the target protein occupancy of the remaining ligand must be significantly less than the target protein occupancy of the starting ligand, and preferably the target protein occupancy of the remaining ligand is very small and negligible. In the case of a high activity ligand at high test concentrations, for example 10,000 times the equilibrium dissociation constant value (Kd), the remaining ligand may result in a false positive strong binding endurance test result. In this case, a more thorough extracellular ligand clearance should be used. Order, for example 3, 4, 5 or more cleanings.
优选地,测定细胞内配体靶蛋白相互作用持久力的方法,其步骤3中收集细胞的时间点至少为8个时间点,相邻时间点之间的间隔根据配体靶蛋白复合物的解离速率决定。Preferably, the method for determining the endurance of the ligand interaction of the ligand in the cell, wherein the time point of collecting the cells in the step 3 is at least 8 time points, and the interval between adjacent time points is based on the solution of the ligand target protein complex The rate of departure is determined.
本发明步骤3中,收集去除细胞外配体后不同时间点的细胞,代表了细胞内配体靶蛋白的解离反应的不同阶段。解离反应的时间点的数目可以是,但不局限于,1,2,3,4,5,6,7,8,9,10,11,20或更多,而较好的是包括至少8个数据点。不同时间点之间的时间间隔根据配体靶蛋白复合物的解离速率决定,可以是1,2,3,5,10,20,30,60分钟;也可以是2,3,4,6,12,24,48小时或更长时间。此外,本发明的方法还包括收集去除细胞外配体前的细胞,代表了本解离反应中配体靶蛋白相互作用的最大起始水平。对于一个细胞解离速率快的配体,去除细胞外配体后,细胞内配体靶蛋白复合物将快速的解离,在此种情况下应在解离反应的早期阶段设置更多的数据点,尤其是分析去除细胞外配体前的细胞。对于一个细胞解离速率慢的配体,去除细胞外配体后,配体靶蛋白复合物将缓慢的解体,在此种情况下应考虑合适的时间间隔设置足够的数据点以覆盖整个解离反应过程。除以上细胞样品外,在一些情况下为了计算细胞内配体结合的靶蛋白的水平和确定细胞内配体靶蛋白占有率的动力学,本发明的方法还可进一步包括阴性对照和阳性对照。阳性对照较好的是用配体处理后但未去除配体的细胞,具有已知的细胞活性水平或已知的靶蛋白占有率,而更好的是用相同试验浓度的配体处理后但未去除的细胞,代表了此试验条件下配体结合的靶蛋白的最大水平。阴性对照反映了此试验条件下的噪音水平,可以是没有配体处理的细胞,而较好的是用前述所说的剩余的配体处理的细胞。此外,阴性对照也可是去除细胞外配体后足够长的时间直到细胞内配体靶蛋白复合物完全解离的细胞。In step 3 of the present invention, cells at different time points after removal of the extracellular ligand are collected, representing different stages of the dissociation reaction of the intracellular ligand target protein. The number of time points of the dissociation reaction may be, but not limited to, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 20 or more, and preferably includes at least 8 data points. The time interval between different time points is determined according to the dissociation rate of the ligand target protein complex, which may be 1, 2, 3, 5, 10, 20, 30, 60 minutes; or 2, 3, 4, 6 , 12, 24, 48 hours or more. In addition, the methods of the invention also include collecting cells prior to removal of the extracellular ligand, representing the maximum initial level of ligand target protein interaction in the dissociation reaction. For a ligand with a fast cell dissociation rate, the intracellular ligand target protein complex will be rapidly dissociated after removal of the extracellular ligand, in which case more data should be set in the early stages of the dissociation reaction. Point, especially the analysis of cells before removal of extracellular ligands. For a ligand with a slow cell dissociation rate, the ligand target protein complex will slowly disintegrate after removal of the extracellular ligand, in which case sufficient data points should be set to cover the entire dissociation at appropriate intervals. reaction process. In addition to the above cell samples, in some cases, to calculate the level of intracellular ligand-bound target protein and to determine the kinetics of intracellular ligand target protein occupancy, the methods of the invention may further comprise a negative control and a positive control. The positive control is preferably a cell treated with a ligand but not having a ligand removed, having a known level of cell activity or a known target protein occupancy, and more preferably treated with the same test concentration of the ligand but Unremoved cells represent the maximum level of ligand-bound target protein under this assay condition. The negative control reflects the level of noise under this test condition, which may be cells without ligand treatment, and is preferably cells treated with the remaining ligands described above. In addition, the negative control may also be a cell that is sufficiently long after removal of the extracellular ligand until the intracellular ligand target protein complex is completely dissociated.
优选地,测定细胞内配体靶蛋白相互作用持久力的方法,其步骤4中对细胞进行加热处理的温度高于配体未结合靶蛋白的起始熔解温度并 且低于配体结合靶蛋白的最终熔解温度。Preferably, the method for determining the endurance of the interaction of the ligand protein in the cell, wherein the temperature at which the cells are heat treated in step 4 is higher than the initial melting temperature of the ligand not bound to the target protein and And below the final melting temperature of the ligand binding target protein.
本发明步骤4中,用细胞热转移技术检测去除细胞外配体后不同时间点的细胞内配体结合的靶蛋白的水平。加热细胞到某一温度并持续一段时间,此时配体未结合的靶蛋白和配体结合的靶蛋白具有不同的热稳定性,通常是配体结合的靶蛋白比未结合的靶蛋白有更高的热熔解温度。加热温度可以是高于配体未结合的靶蛋白的起始熔解温度但低于配体结合的靶蛋白的最终熔解温度区间中的一个温度。加热温度最好的是在阴性对照和样品中可溶靶蛋白具有最显著差异的温度。加热时间可以是任意的时间范围,例如0.5,1,2,3或5分钟,只要在此种情况下阴性对照和样品中可溶靶蛋白的水平有显著的差异,而更好的是有最大程度的差异。加热的仪器包括PCR仪,孵化器,水浴锅等,只要可以加热细胞到特定的温度。In step 4 of the present invention, the level of the intracellular ligand-bound target protein at different time points after removal of the extracellular ligand is detected by a cell thermal transfer technique. Heating the cells to a certain temperature for a period of time, when the ligand-unbound target protein and the ligand-bound target protein have different thermostability, usually the ligand-bound target protein is more than the unbound target protein. High heat melting temperature. The heating temperature may be a temperature above the initial melting temperature of the unbound target protein of the ligand but below the final melting temperature interval of the ligand-bound target protein. The heating temperature is preferably the temperature at which the soluble target protein has the most significant difference in the negative control and sample. The heating time can be any time range, such as 0.5, 1, 2, 3 or 5 minutes, as long as the level of soluble target protein in the negative control and the sample is significantly different in this case, and it is better to have the largest The difference in degree. Heating instruments include PCR machines, incubators, water baths, etc., as long as the cells can be heated to a specific temperature.
本发明步骤5中细胞裂解的方法可以是任意一种能够裂解细胞释放细胞内蛋白的方法。细胞裂解的方法可以是超声破碎法,较好的是包含1,2或3次循环的冻融法,而更好的是用含有离子型或非离子型的变性剂或两性分子的细胞裂解液裂解细胞,尤其是提取膜蛋白,例如50mM Tris-HCl pH7.5,150mM NaCl,1%NP-40。由于细胞裂解的过程中,释放的蛋白酶可降解可溶的靶蛋白,所以细胞裂解液中还进一步含有蛋白酶抑制剂,酶的辅因子如Mg2+,或氧化还原剂如DTT,用以稳定和保护靶蛋白。为了方便检测可溶的和不可溶的靶蛋白,可先用一些分离技术分离可溶的和不可溶的蛋白组分。蛋白分离技术可以是任何一种能够分离可溶的和不可溶的蛋白组分的方法。蛋白分离技术较好的是离心分离,上清是可溶的蛋白组分,沉淀是不可溶的蛋白组分。过滤分离也是较好的,滤液是可溶的蛋白组分,残渣是不可溶的蛋白组分。此外也可用靶蛋白特异性抗体亲和纯化的方法分离可溶的靶蛋白。The method of cell lysis in the step 5 of the present invention may be any method capable of lysing cells to release intracellular proteins. The cell lysis method may be a sonication method, preferably a freeze-thaw method comprising 1, 2 or 3 cycles, and more preferably a cell lysate containing an ionic or non-ionic denaturing agent or amphiphilic molecule. Cells are lysed, especially membrane proteins, such as 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% NP-40. Since the released protease can degrade the soluble target protein during cell lysis, the cell lysate further contains a protease inhibitor, an enzyme cofactor such as Mg 2+ , or a redox agent such as DTT for stabilization and Protect the target protein. To facilitate the detection of soluble and insoluble target proteins, soluble and insoluble protein components can be separated by some separation technique. The protein separation technique can be any method capable of separating soluble and insoluble protein components. The protein separation technique is preferably centrifugal separation, the supernatant is a soluble protein component, and the precipitate is an insoluble protein component. Filtration separation is also preferred, the filtrate is a soluble protein component and the residue is an insoluble protein component. In addition, the soluble target protein can also be isolated by affinity purification of the target protein-specific antibody.
优选地,测定细胞内配体靶蛋白相互作用持久力的方法,其步骤6中靶蛋白的定量分析方法包括抗体技术,质谱法,靶蛋白的生物活性或融合标签的酶活性。 Preferably, the method for determining the endurance of the interaction of the ligand protein in the cell, the method for quantitatively analyzing the target protein in the step 6 comprises antibody technology, mass spectrometry, biological activity of the target protein or enzymatic activity of the fusion tag.
本发明步骤6中,分析可溶的或不可溶的蛋白组分,确定其中可溶的或不可溶的靶蛋白的量。测量不可溶的靶蛋白,需先溶解不可溶的蛋白组分。所以较好的是测量可溶的靶蛋白。靶蛋白的定量方法包括质谱法,例如orbitrap或离子阱;应用靶蛋白或融合标签特异的抗体法,例如免疫印迹,ELISA;以及利用靶蛋白的生物活性或融合标签的酶活性进行定量分析。靶蛋白的定量方法最好的是用高通量的可直接检测细胞裂解液中可溶的靶蛋白的水平的方法,例如alpha扫描,荧光共振能量转移,荧光偏振。In step 6 of the present invention, the soluble or insoluble protein component is analyzed to determine the amount of target protein in which it is soluble or insoluble. To measure an insoluble target protein, it is necessary to first dissolve the insoluble protein component. It is therefore preferred to measure soluble target proteins. Quantitative methods for target proteins include mass spectrometry, such as orbitrap or ion traps; application of target protein or fusion tag-specific antibody methods, such as immunoblotting, ELISA; and quantitative analysis using the biological activity of the target protein or the enzymatic activity of the fusion tag. The best method for quantifying target proteins is to use high-throughput methods for directly detecting the level of soluble target proteins in cell lysates, such as alpha scanning, fluorescence resonance energy transfer, and fluorescence polarization.
优选地,测定细胞内配体靶蛋白相互作用持久力的方法,其步骤6还包括,分析可溶的靶蛋白,确定细胞内配体结合的靶蛋白的水平,以解离时间为函数构建细胞内配体靶蛋白相互作用的解离曲线,计算细胞内配体靶蛋白相互作用的解离速率常数,滞留时间和解离半衰期。Preferably, the method for determining the endurance of the interaction of the ligand protein in the cell, the step 6 further comprises: analyzing the soluble target protein, determining the level of the target protein bound by the ligand in the cell, and constructing the cell as a function of the dissociation time. The dissociation curve of the internal ligand target protein interaction calculates the dissociation rate constant, residence time and dissociation half-life of the ligand ligand protein interaction in the cell.
根据本发明的方法,分析可溶的蛋白组分,测量其中可溶的靶蛋白,确定细胞内配体结合的靶蛋白的量,构建细胞内配体靶蛋白相互作用的解离曲线。样品细胞中比代表噪音水平的阴性对照中检测到更高水平的可溶靶蛋白,意味着在去除细胞外配体后该时间点配体仍然结合在细胞内靶蛋白上。配体结合的靶蛋白的水平等于样品中与阴性对照中可溶的靶蛋白的差值,初步反映了细胞内配体靶蛋白相互作用的持久力的强度。此外,某些加热温度下配体非结合的靶蛋白可完全解折叠而沉淀,在此种情况下,阴性对照可以不是必须的,可溶的靶蛋白的水平可直接反映细胞内配体结合的靶蛋白的水平。用去除细胞外配体后不同时间点的细胞内配体结合的靶蛋白的量以解离时间为函数,可构建细胞内配体靶蛋白相互作用的解离曲线。根据细胞内配体靶蛋白的解离曲线可确定细胞内配体靶蛋白相互作用的解离速率和滞留时间,定量的描述细胞内配体靶蛋白相互作用的持久力。然而本发明发现细胞内配体靶蛋白的解离反应也可包括多个具有显著不同的解离速率的解离阶段,所以在此种情况下应用传统的解离速率和滞留时间很难准确的比较细胞内配体靶蛋白相互作用的持久力(实施例1和2)。为了解决这个问题,本发明的方法进 一步包含根据细胞内配体靶蛋白的解离曲线确定细胞内50%配体靶蛋白复合物解离所需要的时间,称为细胞内配体靶蛋白的解离半衰期,可用于描述细胞内配体靶蛋白相互作用的持久力。相似的也可确定不同百分比的配体靶蛋白复合物解离所需要的时间,例如10%,20%,30%,40%,用于更精确的描述细胞内配体靶蛋白相互作用的持久力。According to the method of the present invention, the soluble protein component is analyzed, the target protein soluble therein is measured, the amount of the target protein bound by the intracellular ligand is determined, and the dissociation curve of the intracellular ligand target protein interaction is constructed. A higher level of soluble target protein was detected in the sample cells than in the negative control representing the noise level, meaning that the ligand still binds to the intracellular target protein at this time point after removal of the extracellular ligand. The level of ligand-bound target protein is equal to the difference between the sample and the soluble target protein in the negative control, initially reflecting the strength of the endurance of the intracellular ligand target protein interaction. In addition, the ligand-unbound target protein at some heating temperatures can be completely unfolded and precipitated. In this case, a negative control may not be necessary, and the level of soluble target protein directly reflects the intracellular ligand binding. The level of the target protein. The dissociation curve of intracellular ligand target protein interactions can be constructed as a function of the dissociation time as a function of the dissociation time by removing the amount of target protein bound by the intracellular ligand at different time points after removal of the extracellular ligand. The dissociation rate and retention time of intracellular ligand target protein interactions can be determined based on the dissociation curve of the intracellular ligand target protein, quantitatively describing the endurance of the intracellular ligand target protein interaction. However, the present inventors have found that the dissociation reaction of the intracellular ligand target protein may also include a plurality of dissociation phases having significantly different dissociation rates, so that it is difficult to accurately apply the conventional dissociation rate and residence time in this case. The persistence of intracellular ligand target protein interactions was compared (Examples 1 and 2). In order to solve this problem, the method of the present invention advances One step comprises determining the time required for dissociation of 50% of the ligand target protein complex in the cell according to the dissociation curve of the intracellular ligand target protein, which is called the dissociation half-life of the intracellular ligand target protein, and can be used to describe intracellular distribution. The persistence of body target protein interactions. Similarly, the time required for dissociation of different percentages of ligand target protein complexes can also be determined, for example 10%, 20%, 30%, 40%, for more precise description of the persistence of intracellular ligand target protein interactions. force.
优选地,测定细胞内配体靶蛋白相互作用持久力的方法,其步骤6还包括确定去除细胞外配体后不同时间点的靶蛋白占有率,构建细胞内配体靶蛋白占有率的动力学曲线,计算高于最低有效靶蛋白占有率的配体的治疗持续时间范围。Preferably, the method for determining the endurance of the interaction of the ligand protein in the cell, the step 6 further comprises determining the target protein occupancy rate at different time points after removing the extracellular ligand, and constructing the kinetics of the ligand protein occupancy rate in the cell. The curve calculates the duration of treatment of the ligand above the lowest effective target protein occupancy.
配体与细胞内靶蛋白具有强的结合持久力,意味着该配体具有产生持久的细胞内药理作用的潜力,然而在一些情况下例如不同的给药剂量即使相同的配体靶蛋白的结合持久力,仍可导致不同的细胞内药理作用的持续时间。为了确定不同的情况下去除细胞外配体后细胞内药理活性的持续时间,本发明的方法进一步包含依据阳性对照中已知的配体靶蛋白占有率和细胞内配体结合的靶蛋白的水平,计算去除细胞外配体后不同时间点的细胞内配体靶蛋白占有率的水平,以此构建去除细胞外配体后细胞内配体靶蛋白占有率的动力学曲线,反映了在此种情况下去除细胞外配体后细胞内配体靶蛋白占有率与解离时间的关系。可依据靶蛋白的生物学特性和致病机制,确定能产生细胞内药理作用的最低的有效靶蛋白占有率。在细胞内配体靶蛋白占有率的动力学曲线中高于最低的有效配体靶蛋白占有率的时间范围,就是在此种情况下细胞外配体消除后细胞内药理作用可以持续的时间范围,本发明称为配体的“治疗持续时间范围”(Therapeutic duration time range)。在不同的情况下可有不同的细胞内配体靶蛋白占有率的动力学曲线,也就会有不同的配体的细胞内药理作用的持续时间范围。当解离反应起始的靶蛋白占有率为100%时,细胞内配体靶蛋白的解离曲线直接等于细胞内配体靶蛋白占有率的动力学曲线,在此种情况下配体的治疗持续时间范围也可从细胞内配体靶蛋白的解离曲线上确定。 Ligand has strong binding endurance with intracellular target proteins, meaning that the ligand has the potential to produce long-lasting intracellular pharmacological effects, however in some cases, for example, different doses of administration, even the same ligand target protein binding Persistence can still lead to different intracellular pharmacological effects over time. In order to determine the duration of intracellular pharmacological activity after removal of the extracellular ligand under different conditions, the method of the invention further comprises determining the occupancy of the ligand target protein and the level of the target protein bound by the intracellular ligand in the positive control. Calculate the level of intracellular ligand target protein occupancy at different time points after removal of the extracellular ligand, thereby constructing a kinetic curve of the intracellular ligand target protein occupancy after removal of the extracellular ligand, reflecting this In the case of removal of extracellular ligands, the relationship between intracellular ligand target protein occupancy and dissociation time. The lowest effective target protein occupancy that produces intracellular pharmacology can be determined based on the biological properties of the target protein and the pathogenic mechanism. The time range in which the intracellular ligand target protein occupancy kinetic curve is higher than the lowest effective ligand target protein occupancy rate is the time range in which the intracellular pharmacological action can be sustained after the extracellular ligand is eliminated. The invention is referred to as the "Therapeutic duration time range" of the ligand. In different cases, there may be different kinetic curves of the intracellular ligand target protein occupancy, and there may be different durations of intracellular pharmacological action of the ligand. When the target protein occupancy rate of the dissociation reaction is 100%, the dissociation curve of the intracellular ligand target protein is directly equal to the kinetic curve of the intracellular ligand target protein occupancy rate, in which case the treatment of the ligand The duration range can also be determined from the dissociation curve of the intracellular ligand target protein.
优选地,测定细胞内配体靶蛋白相互作用持久力的方法,还包括细胞内配体靶蛋白相互作用的解离反应包括去除细胞外配体期间的解离反应和去除细胞外配体后的解离反应。Preferably, the method for determining the persistence of interaction of a ligand protein in a cell, and the dissociation reaction of an intracellular ligand target protein interaction comprises dissociation reaction during removal of the extracellular ligand and removal of the extracellular ligand Dissociation reaction.
根据本发明的方法,发现细胞内配体靶蛋白的解离反应可在去除细胞外配体的过程中明显的发生,在此种情况下如果忽略去除细胞外配体期间的解离反应,可能会导致对评价细胞内配体靶蛋白的结合持久力的强度产生错误的结论。为了确定去除细胞外配体期间的解离反应,本发明的方法还进一步包括分析去除细胞外配体前的细胞,作为一种阳性对照,代表了在该解离试验中配体结合的靶蛋白的最大起始水平,与解离反应第一个时间点样品相比较,可确定在去除细胞外配体期间细胞内配体靶蛋白复合物解离的程度。如实施例1所示,如果未分析去除细胞外配体前的细胞而忽略去除细胞外配体期间的解离反应,可导致将解离反应的第二阶段误认为是整个细胞内配体靶蛋白的解离反应,而得出假阳性的强的结合持久力的结论。然而考虑去除细胞外配体期间的解离反应后,发现细胞内大部分配体在去除细胞外配体期间已从靶蛋白上解离,展示了一个快的解离速率,相反的是仅有少量的配体缓慢的从靶蛋白上解离。综合考虑包括去除细胞外配体期间的整个解离反应后可确定该检测配体分子与细胞内靶蛋白实际上是有一个弱的结合持久力。为了准确的评价细胞内配体靶蛋白的结合持久力,应该考虑应用一个完整的细胞内配体靶蛋白的解离反应,包括去除细胞外配体期间的解离反应和去除细胞外配体后的解离反应。According to the method of the present invention, it is found that the dissociation reaction of the intracellular ligand target protein can be apparently occurred in the process of removing the extracellular ligand, in which case it is possible to ignore the dissociation reaction during removal of the extracellular ligand, possibly This leads to erroneous conclusions about the strength of the binding endurance of the ligand target protein in the cell. To determine the dissociation reaction during removal of the extracellular ligand, the method of the invention further comprises analyzing the cells prior to removal of the extracellular ligand as a positive control representing the ligand-bound target protein in the dissociation assay. The maximum initial level, as compared to the first time point sample of the dissociation reaction, determines the extent to which the intracellular ligand target protein complex dissociates during removal of the extracellular ligand. As shown in Example 1, if the cells before removal of the extracellular ligand are not analyzed and the dissociation reaction during removal of the extracellular ligand is neglected, the second stage of the dissociation reaction may be mistaken for the entire intracellular ligand target. The dissociation reaction of the protein results in a strong binding endurance of the false positive. However, after considering the dissociation reaction during removal of the extracellular ligand, it was found that most of the ligands in the cell had dissociated from the target protein during removal of the extracellular ligand, demonstrating a fast dissociation rate, and conversely only A small amount of the ligand slowly dissociates from the target protein. Considering the entire dissociation reaction during removal of the extracellular ligand, it can be determined that the test ligand molecule actually has a weak binding endurance with the intracellular target protein. In order to accurately assess the binding endurance of intracellular ligand target proteins, consideration should be given to the application of a complete intracellular ligand target protein dissociation reaction, including removal of extracellular ligands during dissociation and removal of extracellular ligands. Dissociation reaction.
如上所述,不同于重组表达纯化的靶蛋白,细胞内靶蛋白并不是完全均一的成分,实际上包含不同构象的靶蛋白,不同翻译后修饰的靶蛋白,不同亚细胞器定位的靶蛋白,以及与不同的结合伙伴相互作用的靶蛋白,不同状态的靶蛋白与配体分子可能有不同的快的或慢的解离速率。此外,在解离反应的过程中,细胞内配体结合的或非结合的靶蛋白可能会降解,新的非结合的靶蛋白可能会合成,细胞内靶蛋白的降解与合成可减弱细胞内配体靶蛋白的结合持久力。相似的是细胞增殖也可导致减 弱细胞内配体靶蛋白相互作用的持久力。以上这些机制可能会有助于解释本发明发现的细胞内配体靶蛋白的多阶段的解离反应。As described above, unlike recombinantly expressed and purified target proteins, intracellular target proteins are not completely homogeneous components, and actually contain target proteins of different conformations, different post-translationally modified target proteins, different subcellular organelle target proteins, and Target proteins that interact with different binding partners may have different fast or slow dissociation rates for target proteins and ligand molecules in different states. In addition, during the dissociation reaction, intracellular ligand-bound or unbound target proteins may be degraded, new unbound target proteins may be synthesized, and degradation and synthesis of intracellular target proteins may attenuate intracellular distribution. The binding endurance of the body target protein. Similar to cell proliferation can also lead to reduction The persistence of ligand-protein interactions in weak cells. These mechanisms may be helpful in explaining the multi-stage dissociation reaction of the intracellular ligand target protein found in the present invention.
由于细胞内配体靶蛋白的解离试验可能会持续一个长的时间范围,尤其是对于一些解离速率慢的配体,例如48或96小时。解离反应期间细胞会继续生长,所以为了准确的比较不同时间点的细胞内配体结合的靶蛋白的水平,本发明的方法还进一步包含归一化处理。归一化处理的标准可以是根据可溶的细胞裂解液的蛋白浓度或者根据样品的细胞数目。细胞的增殖会降低细胞内配体靶蛋白复合物的平均水平,相应的会导致减弱细胞内配体靶蛋白相互作用的持久力,然而体内的细胞增殖速率通常会显著低于体外的细胞增值速率,在此种情况下可导致由于体外快的细胞增殖速度而低估配体靶蛋白相互作用的持久力,所以为了在接近体内的情况下评价细胞内配体靶蛋白相互作用的持久力需排除体外高的细胞增殖速率的影响,可采用相同取样体积的细胞,例如1ml或1孔,作为归一化标准。在此种情况下不同的时间点的样品可能含有不同数目的细胞但拥有相同的起始水平的配体靶蛋白复合物,在一定程度上也可理解此种情况下细胞增殖速率为0(实施例3)。此种归一化标准尤其适合体内增殖速率慢的细胞,可避免由于体外细胞增殖导致低估配体靶蛋白相互作用的持久力,更好的反映体内的配体靶蛋白相互作用的持久力。Dissociation assays due to intracellular ligand target proteins may persist for a long period of time, especially for ligands with slow dissociation rates, such as 48 or 96 hours. The cells continue to grow during the dissociation reaction, so in order to accurately compare the levels of intracellular ligand-bound target proteins at different time points, the method of the present invention further includes normalization. The standard for normalization can be based on the protein concentration of the soluble cell lysate or on the number of cells in the sample. Proliferation of cells reduces the average level of ligand-target protein complexes in cells, which in turn leads to a decrease in the persistence of ligand-protein interactions in cells. However, the rate of cell proliferation in vivo is usually significantly lower than the rate of cell proliferation in vitro. In this case, the survival of the ligand target protein interaction is underestimated due to the rapid cell proliferation rate in vitro, so in order to evaluate the endurance of the ligand interaction of the intracellular ligand in the case of proximity to the body, it is necessary to exclude the in vitro For high cell proliferation rates, cells of the same sample volume, such as 1 ml or 1 well, can be used as normalization criteria. In this case, samples at different time points may contain different numbers of cells but possess the same initial level of ligand target protein complex. To some extent, it is also understood that the cell proliferation rate is 0 in this case. Example 3). This normalization standard is especially suitable for cells with slow proliferation rate in vivo, which can avoid the insufficiency of ligand cell target interaction due to in vitro cell proliferation, and better reflect the endurance of ligand target protein interaction in vivo.
本发明的方法可用于新药研发,确定不同化合物的细胞内靶蛋白的结合持久力,依据靶蛋白的生物学特性和病理学特征选择合适的结合持久力的化合物作为苗头分子,先导化合物,候选药物或药物。强持久力的化合物比弱持久力的化合物在相同的解离时间点会有更高水平的可溶的靶蛋白,也会有更慢的细胞内解离速率和更长的细胞内滞留时间,尤其是拥有更长的治疗持续时间范围。化合物的结合持久力参数可用于指导先导化合物的优化,设计合成具有期望的结合持久力的药物分子。此外,当面对多个具有相同的细胞活性的化合物时,细胞内化合物靶蛋白相互作用的持久力可作为一种新的参数,从时间的角度进一步对化合物进行排序和区分,有助于选择最优的活性分子。 The method of the invention can be used for the development of new drugs, determining the binding endurance of intracellular target proteins of different compounds, selecting suitable binding endurance compounds as seed molecules, lead compounds, drug candidates according to the biological characteristics and pathological characteristics of the target protein. Or drugs. Strongly potent compounds have higher levels of soluble target protein at the same dissociation time point than weakly durable compounds, as well as slower intracellular dissociation rates and longer intracellular residence times. In particular, it has a longer duration of treatment. The binding endurance parameter of the compound can be used to guide the optimization of the lead compound and to design and synthesize a drug molecule having the desired binding endurance. In addition, when faced with multiple compounds with the same cellular activity, the persistence of intracellular compound target protein interactions can be used as a new parameter to further sort and distinguish compounds from a time perspective, which is helpful for selection. The most active molecule.
本发明的方法可用于比较化合物与细胞内不同靶蛋白的结合持久力,从时间的角度评价化合物细胞内靶蛋白的选择性,称为时间的细胞内靶蛋白的选择性。活性分子通常可与细胞内多个蛋白相互作用,其中负责药理活性的称为靶蛋白,其它的称为脱靶蛋白。活性分子和脱靶蛋白的结合有时可能会导致有害的副作用,所以药物的靶蛋白选择性是先导化合物优化阶段的一个重要目标。传统的靶蛋白选择性主要是从热力学的角度比较活性分子与靶蛋白的和与脱靶蛋白的亲和力。互补于传统的靶蛋白选择性,本发明的时间的细胞内靶蛋白选择性是指依据细胞内靶蛋白占有率和脱靶蛋白占有率的动力学,比较相同时间点的靶蛋白占有率和脱靶蛋白占有率,或比较细胞内靶蛋白的滞留时间和脱靶蛋白的滞留时间,从动力学的角度确定活性分子的细胞内靶蛋白选择性。面对与靶蛋白和脱靶蛋白有相同的亲和力的活性分子,可用本发明的时间的细胞内靶蛋白选择性从动力学的角度进一步评价活性分子的靶蛋白选择性。相似的,本发明的方法也可用于比较活性分子与野生型和突变体的靶蛋白的结合持久力,从时间的角度确定活性分子对于野生型和突变体的选择性。The method of the present invention can be used to compare the binding endurance of a compound to a different target protein in a cell, and to evaluate the selectivity of a target protein in a compound cell from a time perspective, which is called the selectivity of a target protein in time. Active molecules usually interact with multiple proteins in the cell, of which the target protein is responsible for pharmacological activity, and the other is called off-target protein. The binding of active molecules to off-target proteins can sometimes lead to deleterious side effects, so the target protein selectivity of the drug is an important goal in the optimization phase of the lead compound. The traditional target protein selectivity is mainly to compare the affinity of the active molecule to the target protein and the off-target protein from a thermodynamic point of view. Complementing the traditional target protein selectivity, the intracellular target protein selectivity of the present invention refers to the kinetics of target protein occupancy and off-target protein occupancy in the cell, comparing target protein occupancy and off-target protein at the same time point. The occupancy rate, or the retention time of the target protein in the cell and the retention time of the off-target protein, determine the intracellular target protein selectivity of the active molecule from the kinetic point of view. In the presence of an active molecule having the same affinity as the target protein and the off-target protein, the target protein selectivity of the active molecule can be further evaluated from the kinetic point of view using the intracellular target protein of the present invention. Similarly, the methods of the invention can also be used to compare the binding endurance of an active molecule to a wild-type and mutant target protein, determining the selectivity of the active molecule for wild-type and mutant from a time perspective.
本发明的方法可用于比较活性化合物在不同的细胞中与靶蛋白相互作用的持久力,从时间的角度评价活性分子的细胞选择性。靶蛋白可分布于体内多个组织和器官,例如心,肺,肾,肝,脑等,其中与活性分子的药理活性相关的组织称为靶组织,活性分子与其它组织中的靶蛋白相互作用可能会导致不良的副作用。虽然不同组织中靶蛋白有相同的氨基酸序列,但是不同的细胞环境可能会导致不同的活性分子靶蛋白相互作用的持久力。本发明从时间的角度评价活性分子在不同的组织细胞中的选择性有助于解释药物的作用机制,设计合理的给药方案,降低脱靶组织相关的药物副作用。相似的,本发明的方法可用于比较活性化合物在不同的肿瘤细胞中与靶蛋白相互作用的持久力,从时间的角度评价活性分子的肿瘤细胞的选择性,有助于预测药物在不同肿瘤中的疗效,设计个性化的给药方案。 The method of the present invention can be used to compare the persistence of an active compound interacting with a target protein in different cells, and to evaluate the cell selectivity of the active molecule from a time perspective. The target protein can be distributed in multiple tissues and organs in the body, such as heart, lung, kidney, liver, brain, etc., wherein the tissue related to the pharmacological activity of the active molecule is called a target tissue, and the active molecule interacts with a target protein in other tissues. May cause undesirable side effects. Although target proteins in different tissues have the same amino acid sequence, different cellular environments may result in the persistence of interactions between different active molecular target proteins. The present invention evaluates the selectivity of active molecules in different tissue cells from a time perspective to help explain the mechanism of action of the drug, design a rational dosing regimen, and reduce the side effects associated with off-target tissue. Similarly, the method of the present invention can be used to compare the persistence of active compounds interacting with target proteins in different tumor cells, and to evaluate the selectivity of tumor cells of active molecules from a time perspective, and to help predict drugs in different tumors. The efficacy of the design of personalized dosing regimens.
本发明的方法可用于预测设计在动物或人体内最有效的给药方案,尤其是根据药物的治疗持续时间范围预测给药的时间间隔。给药方案包括给药剂量和给药间隔,对生物活性分子在动物和人体内的疗效和毒性具有显著的影响。给药的时间间隔通常是根据该化合物的药代动力学确定,认为药物的血药浓度一旦低于治疗浓度范围,药物的药理活性将立即终止。然而本发明已经证明某些化合物即使去除细胞外化合物后24小时仍能以约50%靶蛋白占有率与细胞内靶蛋白相互作用(实施例2&3),意味着该化合物的细胞活性即使去除细胞外化合物后24小时仍可持续存在。应用药物的药代动力学特性结合本发明的药物的治疗持续时间范围预测给药的时间间隔将会是更加科学与合理,因此本发明的方法有助于设计最佳的给药方案,通过慢的细胞解离速率维持持续的靶蛋白占有率而获得疗效,通过合适的给药间隔而减弱脱靶蛋白相关的毒副作用,以此得到药物最大的治疗窗。例如某一化合物在体内可2小时内完全清除,然而在此种情况下有一个24小时的治疗持续时间范围。根据药代动力学特性,该化合物需要每天多次给药,然而考虑该化合物的治疗持续时间范围后,可降低为一天一次的给药方案。The methods of the present invention can be used to predict the most effective dosing regimen for design in an animal or human, particularly in predicting the time interval of administration based on the duration of treatment of the drug. Dosing regimens, including dosing and dosing intervals, have a significant impact on the efficacy and toxicity of bioactive molecules in animals and humans. The time interval for administration is usually determined based on the pharmacokinetics of the compound, and it is considered that once the blood concentration of the drug is below the therapeutic concentration range, the pharmacological activity of the drug will be immediately terminated. However, the present inventors have demonstrated that certain compounds can interact with intracellular target proteins at about 50% target protein occupancy even after removal of extracellular compounds (Examples 2 & 3), meaning that the cellular activity of the compound is even if the cells are removed. The compound is still present 24 hours after the compound. It is more scientific and rational to predict the time interval of administration in combination with the pharmacological kinetic properties of the drug in combination with the therapeutic duration of the drug of the present invention, and thus the method of the present invention facilitates the design of an optimal dosing regimen, The rate of cell dissociation maintains a sustained target protein occupancy and achieves therapeutic efficacy, and the toxic side effects associated with the off-target protein are attenuated by appropriate dosing intervals, thereby obtaining the largest therapeutic window of the drug. For example, a compound can be completely eliminated within 2 hours in the body, however in this case there is a 24 hour treatment duration range. Depending on the pharmacokinetic properties, the compound requires multiple administrations per day, however, considering the therapeutic duration of the compound, the once-daily dosing regimen can be reduced.
与现有技术相比,本发明的有益效果如下:Compared with the prior art, the beneficial effects of the present invention are as follows:
本发明的方法不需要使用荧光探针分子,因此本发明的方法没有探针分子带来的干扰效应可以准确的测量细胞内配体靶蛋白相互作用的持久力;本发明的方法没有探针分子带来的延迟效应可以完整的记录细胞内配体靶蛋白的解离反应。其次,本发明的方法不需要构建融合蛋白,可检测天然蛋白,尤其是直接来自病人的。最后,本发明的方法是一个通用的方法,可分析绝大部分的配体和靶蛋白。应用此种方法,发现细胞内配体靶蛋白解离反应要比已报道的数据复杂的多,可包含多个具有显著不同解离速率的解离阶段。最重要的是发现清除细胞外配体后24小时,细胞内配体靶蛋白占有率仍能维持在约60%的水平。因此本发明的方法代表了一种有效的测定细胞内配体靶蛋白相互作用的持久力的方法。 The method of the present invention does not require the use of fluorescent probe molecules, so the method of the present invention can accurately measure the endurance of intracellular ligand target protein interaction without the interference effect brought by the probe molecule; the method of the present invention has no probe molecule The delayed effect can completely record the dissociation reaction of the ligand target protein in the cell. Secondly, the method of the invention does not require the construction of a fusion protein, which can detect native proteins, especially directly from the patient. Finally, the method of the invention is a versatile method for the analysis of the vast majority of ligands and target proteins. Using this approach, it was found that the intracellular ligand target protein dissociation reaction is much more complex than the reported data and can contain multiple dissociation phases with significantly different dissociation rates. Most importantly, the intracellular ligand target protein occupancy was maintained at about 60% 24 hours after removal of the extracellular ligand. Thus the method of the invention represents an effective method for determining the persistence of interactions of target protein proteins within a cell.
附图说明DRAWINGS
图1是Jurkat细胞内Aurora A在缺少(●)和呈现(▲)星形孢菌素的情况下的热熔解曲线图。Figure 1 is a graph of the thermal melting of Aurora A in Jurkat cells in the absence (•) and presence (▲) staurosporine.
图2是Jurkat细胞内Aurora A与星形孢菌素的剂量效应曲线图。Figure 2 is a graph showing the dose response of Aurora A and staurosporine in Jurkat cells.
图3是Jurkat细胞内星形孢菌素Aurora A相互作用的解离曲线图,归一化标准是每个时间点相同数目的细胞。Figure 3 is a dissociation plot of the interaction of staurosporin Aurora A in Jurkat cells, normalized to the same number of cells at each time point.
图4是Jurkat细胞内星形孢菌素Aurora A占有率的动力学曲线图,归一化标准是每个时间点相同数目的细胞。Figure 4 is a kinetic plot of the occupancy of staurosporin Aurora A in Jurkat cells, normalized to the same number of cells at each time point.
图5是Jurkat细胞内PARP-1在缺少(●)和呈现(▲)Talazoparib的情况下的热熔解曲线图。Figure 5 is a graph of the thermal melting of PARP-1 in Jurkat cells in the absence (•) and presence (▲) Talazoparib.
图6是Jurkat细胞内Talazoparib与PARP-1的剂量效应曲线图。Figure 6 is a graph showing the dose response of Talazoparib and PARP-1 in Jurkat cells.
图7是Jurkat细胞内Talazoparib与PARP-1相互作用的解离曲线图,归一化标准是每个时间点相同数目的细胞。Figure 7 is a dissociation plot of Talazoparib interaction with PARP-1 in Jurkat cells, normalized to the same number of cells at each time point.
图8是Jurka细胞内Talazoparib与PARP-1占有率的动力学曲线图,归一化标准是每个时间点相同数目的细胞。Figure 8 is a graph showing the kinetics of Talazoparib and PARP-1 occupancy in Jurka cells, the normalization criterion being the same number of cells at each time point.
图9是Jurkat细胞内Talazoparib与PARP-1相互作用的解离曲线图,归一化标准是每个时间点相同体积的细胞,模拟细胞增殖速度为0的情况。Figure 9 is a dissociation plot of Talazoparib interaction with PARP-1 in Jurkat cells, normalized to the same volume of cells at each time point, simulating a cell proliferation rate of zero.
图10是Jurkat细胞内Talazoparib与PARP-1占有率的动力学曲线,归一化标准是每个时间点相同体积的细胞,模拟细胞增速度为0的情况。Figure 10 is a kinetic curve of Talazoparib and PARP-1 occupancy in Jurkat cells. The normalization criterion is the same volume of cells at each time point, simulating a cell growth rate of zero.
具体实施方式detailed description
下面结合实施例对本发明的技术方案进行详细说明。The technical solution of the present invention will be described in detail below with reference to the embodiments.
实施例1 测定细胞内星形孢菌素Aurora A相互作用的持久力Example 1 Determination of the persistence of intracellular staurosporin Aurora A interaction
本实验的目的是测定Jurkat细胞内星形孢菌素(Staurosporine)与Aurora A相互作用的持久力,发现大部分的星形孢菌素可在去除细胞外星形孢菌素的过程中从细胞内Aurora A上解离,证明细胞内星形孢菌素与Aurora A有一个弱的结合持久力。The purpose of this experiment was to determine the persistence of staurosporine interaction with Aurora A in Jurkat cells and found that most of the staurosporins can be removed from cells during the removal of extracellular staurosporine. Dissociation on Aurora A demonstrates that intracellular staurosporine has a weak binding endurance with Aurora A.
热熔解试验:用RPMI1640培养基37℃5%二氧化碳培养箱中培养 Jurkat细胞。无血清RPMI1640培养基中用10μM星形孢菌素(#19-123MG,Merck KGaA,德国)处理Jurkat细胞半小时,平行的用0.1%DMSO处理的Jurkat细胞作为阴性对照。每个数据点收集1x106细胞重悬在20μL PBS中,用PCR仪(Eppendorf,德国)在37-64℃温度范围内的不同温度下加热三分钟,然后用含有蛋白酶抑制剂(Roche,瑞士)的细胞裂解液(1%NP-40,150mM NaCl,50mM Tris-HCl pH7.5)冰上裂解细胞半小时。离心后,收集上清,用SDS-PAGE和免疫印迹检测Aurora A。相对于最低加热温度下Aurora A的水平,构建有和无星形孢菌素处理的条件下Jurkat细胞内Aurora A的热熔解曲线。Thermal melting test: Jurkat cells were cultured in a RPMI 1640 medium at 37 ° C in a 5% carbon dioxide incubator. Jurkat cells were treated with 10 μM staurosporin (#19-123MG, Merck KGaA, Germany) for half an hour in serum-free RPMI1640 medium, and Jurkat cells treated with 0.1% DMSO in parallel were used as negative controls. 1x10 6 cells were collected from each data point and resuspended in 20 μL of PBS, heated by PCR instrument (Eppendorf, Germany) at different temperatures in the temperature range of 37-64 ° C for three minutes, and then containing protease inhibitors (Roche, Switzerland) The cell lysate (1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5) was lysed on ice for half an hour. After centrifugation, the supernatant was collected and Aurora A was detected by SDS-PAGE and immunoblotting. The thermal melting curve of Aurora A in Jurkat cells under conditions of no treatment with staurosporine was constructed relative to the level of Aurora A at the lowest heating temperature.
剂量效应试验:用不同浓度的从0.1nM到10μM的星形孢菌素在无血清RPMI1640培养基中处理Jurkat细胞1个小时;DMSO浓度为0.1%。每个浓度点取1x106细胞重悬在20μL PBS中,用PCR仪在58℃下加热三分钟。然后用含有蛋白酶抑制剂(Roche,瑞士)的细胞裂解液(1%NP-40,150mM NaCl,50mM Tris-HCl pH7.5)冰上裂解细胞半小时。离心后,收集上清,用SDS-PAGE和免疫印迹检测Aurora A。样品中减去阴性对照中的Aurora A的水平作为星形孢菌素结合的Aurora A的水平,构建细胞内星形孢菌素与Aurora A的剂量效应曲线。Dose-effect assay: Jurkat cells were treated with different concentrations of staurosporine from 0.1 nM to 10 μM in serum-free RPMI 1640 medium for 1 hour; DMSO concentration was 0.1%. 1×10 6 cells were resuspended in 20 μL of PBS at each concentration point, and heated by a PCR machine at 58 ° C for three minutes. The cells were then lysed on ice for half an hour with a cell lysate containing protease inhibitor (Roche, Switzerland) (1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5). After centrifugation, the supernatant was collected and Aurora A was detected by SDS-PAGE and immunoblotting. The level of Aurora A in the negative control was subtracted from the sample as the level of staurosporine-conjugated Aurora A, and a dose-response curve of intracellular staurosporine and Aurora A was constructed.
细胞解离试验:用100nM星形孢菌素在无血清RPMI1640培养基中处理Jurkat细胞半小时,DMSO浓度为0.1%。更换新鲜的含有10%血清的RPMI1640培养基,以此去除细胞外星形孢菌素。然后在不同的时间点收集Jurkat细胞,每个时间点收集1x106细胞,重悬在20μL PBS中,用PCR仪在58℃加热三分钟。然后用含蛋白酶抑制剂(Roche,瑞士)的细胞裂解液(1%NP-40,150mM NaCl,50mM Tris-HCl pH7.5)冰上裂解细胞半个小时。离心后,收集上清,用SDS-PAGE和免疫印迹检测Aurora A。平行的,用去除细胞外星形孢菌素前的Jurkat细胞作为阳性对照,代表了该解离试验的最大起始信号水平和已测定的76%Aurora A占有率(依据以上剂量效应曲线)。用0.1%DMSO处理的Jurkat细胞作为阴性对照,代表了本试验条件下的噪音水平。样品中检测到比阴性对照中更高 水平的Aurora A,意味着去除细胞外星形孢菌素后该时间点星形孢菌素仍与细胞内Aurora A相互作用。通过从样品中减去阴性对照中的Aurora A的水平,作为该样品中星形孢菌素结合的Aurora A的水平,以时间为函数构建细胞内星形孢菌素Aurora A相互作用的解离曲线,确定细胞内星形孢菌素Aurora A相互作用的解离速率,滞留时间和解离半衰期。进一步,依据阳性对照中已知的星形孢菌素Aurora A占有率,确定本试验中细胞内星形孢菌素Aurora A占有率的动力学曲线,确定该试验条件下星形孢菌素的治疗持续时间范围。Cell dissociation assay: Jurkat cells were treated with 100 nM staurosporine in serum-free RPMI 1640 medium for half an hour with a DMSO concentration of 0.1%. Extracellular staurosporine was removed by replacing fresh RPMI1640 medium containing 10% serum. Then collected at various time points Jurkat cells, 1x10 6 per time point were collected cells were resuspended in 20μL PBS and heated at 58 deg.] C using a PCR machine for three minutes. The cells were then lysed on ice for half an hour with a cell lysate containing protease inhibitor (Roche, Switzerland) (1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5). After centrifugation, the supernatant was collected and Aurora A was detected by SDS-PAGE and immunoblotting. Parallel, using Jurkat cells prior to removal of extracellular staurosporine as a positive control, represents the maximum initial signal level for the dissociation test and the 76% Aurora A occupancy measured (based on the above dose-response curve). Jurkat cells treated with 0.1% DMSO as a negative control represent the noise levels under the conditions of this test. A higher level of Aurora A was detected in the sample than in the negative control, meaning that staurosporin still interacted with intracellular Aurora A at this time point after removal of extracellular staurosporine. Dissociation of intracellular staurosporin Aurora A interaction by time as a function of the level of Aurora A in the negative control from the sample as the level of staurosporine-conjugated Aurora A in the sample The curve determines the dissociation rate, retention time and dissociation half-life of intracellular staurosporin Aurora A interaction. Further, based on the known occupancy rate of staurosporin Aurora A in the positive control, the kinetic curve of intracellular staurosporin Aurora A occupancy in this test was determined, and the staurosporin was determined under the test conditions. Duration of treatment.
SDS-PAG&免疫印迹:用SDS-PAGE分离Aurora A蛋白,用Trans-Blot Turbo(Bio-Rad,美国)转印到PVDF膜上,封闭后,用抗Aurora A抗体检测(#14475,Cell Signaling Technology,USA),用CCD相机(GE Healthcare,美国)记录化学发光信号。SDS-PAG & Immunoblotting: Aurora A protein was separated by SDS-PAGE and transferred to PVDF membrane using Trans-Blot Turbo (Bio-Rad, USA), blocked and tested with anti-Aurora A antibody (#14475, Cell Signaling Technology) , USA), the chemiluminescence signal was recorded using a CCD camera (GE Healthcare, USA).
参见图1,该图为Jurkat细胞内Aurora A在缺少(●)和呈现(▲)星形孢菌素的情况下的热熔解曲线图。其中,(●)表示未用星形孢菌素处理的情况下Aurora A的热熔解曲线图;(▲)表示用星形孢菌素处理情况下Aurora A的热熔解曲线图。其中,纵坐标“相对条带强度”是指实施例1中免疫印迹测定的各数据结果的Aurora A发光强度相对于最强Aurora A发光强度的比值。通过图1所示的Aurora A热熔解曲线可知:星形孢菌素处理组比对照组在温度区间55-61℃展示了更高水平的Aurora A,所以可选择58℃作为加热温度用于细胞内星形孢菌素与Aurora A的解离试验。See Figure 1, which is a graph of the thermal melting of Aurora A in Jurkat cells in the absence (•) and presence (▲) staurosporin. Among them, (●) indicates a thermal melting curve of Aurora A in the case of no treatment with staurosporine; (▲) indicates a thermal melting curve of Aurora A in the case of treatment with staurosporine. Here, the ordinate "relative strip intensity" refers to the ratio of the Aurora A luminescence intensity to the strongest Aurora A luminescence intensity of each data result measured by immunoblotting in Example 1. According to the Aurora A thermal melting curve shown in Fig. 1, the staurosporin treatment group exhibited a higher level of Aurora A than the control group at a temperature interval of 55-61 ° C, so that 58 ° C can be selected as the heating temperature for the cells. Dissociation test of endosporin and Aurora A.
参见图2,该图为细胞内Aurora A与星形孢菌素的剂量效应曲线图。如图2所示,细胞内星形孢菌素结合的Aurora A的水平随星形孢菌素浓度(0.1-10000nM范围内)的增加而增加,所以可选择100nM浓度的星形孢菌素,对应Jurkat细胞内76%Aurora A占有率,作为解离试验中星形孢菌素的试验浓度。See Figure 2, which is a graph of the dose response of intracellular Aurora A and staurosporine. As shown in Figure 2, the level of intracellular staurosporin-conjugated Aurora A increases with the concentration of staurosporine (in the range of 0.1-10000 nM), so a concentration of 100 nM staurosporine can be selected. Corresponding to the 76% Aurora A occupancy in Jurkat cells as the test concentration of staurosporin in the dissociation test.
参见图3和图4,图3是Jurkat细胞内星形孢菌素Aurora A相互作用的解离曲线图;图4是Jurkat细胞内星形孢菌素Aurora A占有率的动力学曲线图。归一化标准是每个时间点相同数目的细胞。如图3所示,星形孢菌素结 合的Aurora A的水平随解离时间的增加而非线性降低。应用一个两相解离模型分析Jurkat细胞内星形孢菌素Aurora A的解离曲线(如图3所示)。第一相是一个非常快的解离阶段,发生在去除细胞外星形孢菌素的过程中,比较去除细胞外星形孢菌素前的细胞样品和去除细胞外星形孢菌素后第一个解离时间点样品可发现在约5分钟期间约70%星形孢菌素Aurora A复合物解离,由于非常快的解离速率且有限的数据点,很难准确拟合该相解离速率常数。第二相是一个慢的解离阶段,接下来的36小时内约26%星形孢菌素Aurora A复合物逐渐的解离,该相的解离速率常数为0.042h-1,可推测Jurkat细胞内星形孢菌素Aurora A相互作用的滞留时间约为36小时,或更长一点,然而其解离半衰期仅为约0.1小时。依据阳性对照中76%Aurora A占有率,可将上述细胞内星形孢菌素解离曲线(如图3所示)转换为该试验条件下Jurkat细胞内星形孢菌素Aurora A占有率的动力学曲线(如图4所示),可从此动力学曲线中确定在该试验条件下超过某一Aurora A占有率的时间范围。Referring to Figures 3 and 4, Figure 3 is a dissociation curve of the interaction of staurosporin Aurora A in Jurkat cells; Figure 4 is a kinetic plot of the occupancy of staurosporin Aurora A in Jurkat cells. The normalization criterion is the same number of cells at each time point. As shown in Figure 3, the level of staurosporin-conjugated Aurora A decreased non-linearly with increasing dissociation time. A two-phase dissociation model was used to analyze the dissociation curve of staurosporin Aurora A in Jurkat cells (shown in Figure 3). The first phase is a very fast dissociation phase that occurs during the removal of extracellular staurosporine, comparing cell samples prior to removal of extracellular staurosporine and removal of extracellular staurosporin A dissociation time point sample found that about 70% of the staurosporin Aurora A complex dissociated during about 5 minutes, and it was difficult to accurately fit the phase solution due to the very fast dissociation rate and limited data points. Rate constant. The second phase is a slow dissociation phase, and approximately 26% of the staurosporin Aurora A complex gradually dissociates in the next 36 hours. The dissociation rate constant of this phase is 0.042 h -1 , presumably Jurkat The retention time of intracellular staurosporin Aurora A interaction is about 36 hours, or longer, but its dissociation half-life is only about 0.1 hour. According to the 76% Aurora A occupancy rate in the positive control, the intracellular staurosporin dissociation curve (shown in Figure 3) can be converted to the occupancy of staurosporin Aurora A in Jurkat cells under the test conditions. A kinetic curve (shown in Figure 4) from which the time range over which a certain Aurora A occupancy is exceeded under the test conditions can be determined.
实施例2 测定Jurkat细胞内Talazoparib和PARP-1相互作用的持久力Example 2 Determination of the persistence of Talazoparib and PARP-1 interactions in Jurkat cells
本实验的目的是测定Jurkat细胞内Talazoparib PARP-1相互作用的持久力,发现细胞内Talazoparib与PARP-1有一个强的结合持久力。The purpose of this experiment was to determine the persistence of the Talazoparib PARP-1 interaction in Jurkat cells and found that intracellular Talazoparib has a strong binding endurance with PARP-1.
热熔解试验:用RPMI1640培养基37℃5%二氧化碳培养箱中培养Jurkat细胞。用1μM Talazoparib(Selleckchem,美国)在无血清RPMI1640培养基中处理Jurkat细胞半个小时,平行的用0.1%DMSO处理的细胞作为阴性对照。每个数据点收集1x106细胞重悬在20μL PBS中,用PCR仪(Eppendorf,德国)在37-55℃范围内的不同温度下加热三分钟,然后用含有蛋白酶抑制剂(Roche,瑞士)的细胞裂解液(1%NP-40,150mM NaCl,50mM Tris-HCl pH7.5)冰上裂解细胞半个小时。离心后,收集上清,用SDS-PAGE和免疫印迹检测PARP-1。相对于最低加热温度下PARP-1的水平,构建在有和无Talazoparib处理的条件下细胞内PARP-1的热熔解曲线。Thermal Melting Test: Jurkat cells were cultured in a RPMI 1640 medium at 37 ° C in a 5% carbon dioxide incubator. Jurkat cells were treated with 1 μM Talazoparib (Selleckchem, USA) in serum-free RPMI 1640 medium for half an hour, and cells treated with 0.1% DMSO in parallel served as a negative control. 1x10 6 cells were collected from each data point and resuspended in 20 μL of PBS, heated with a PCR instrument (Eppendorf, Germany) at different temperatures ranging from 37-55 ° C for three minutes, and then containing a protease inhibitor (Roche, Switzerland). Cell lysate (1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5) was lysed on ice for half an hour. After centrifugation, the supernatant was collected, and PARP-1 was detected by SDS-PAGE and immunoblotting. The thermal melting curve of intracellular PARP-1 was constructed in the presence and absence of Talazoparib treatment relative to the level of PARP-1 at the lowest heating temperature.
剂量效应试验:用不同浓度从0.001nM到10μM的Talazoparib在 无血清RPMI1640培养基中处理Jurkat细胞0.5小时,DMSO浓度为0.1%。每个浓度点收集1x106细胞重悬在20μL PBS中,用PCR仪在49℃加热三分钟。然后用含有蛋白酶抑制剂(Roche,瑞士)的细胞裂解液(1%NP-40,150mM NaCl,50mM Tris-HCl pH7.5)冰上裂解细胞半个小时。离心后,收集上清,用SDS-PAGE和免疫印迹检测PARP-1。样品中减去阴性对照中的PARP-1的水平作为细胞内Talazoparib结合的PARP-1的水平,构建Jurkat细胞内Talazoparib与PARP-1的剂量效应曲线。Dose-effect test: Jurkat cells were treated with Talazoparib at different concentrations from 0.001 nM to 10 μM in serum-free RPMI 1640 medium for 0.5 hour at a DMSO concentration of 0.1%. 1×10 6 cells were collected from each concentration point and resuspended in 20 μL of PBS, and heated at 49 ° C for three minutes using a PCR machine. The cells were then lysed on ice for half an hour with a cell lysate containing protease inhibitor (Roche, Switzerland) (1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5). After centrifugation, the supernatant was collected, and PARP-1 was detected by SDS-PAGE and immunoblotting. The level of PARP-1 in the negative control was subtracted from the sample as the level of intracellular Talazoparib-bound PARP-1, and a dose-response curve of Talazoparib and PARP-1 in Jurkat cells was constructed.
细胞解离试验:用100nM Talazoparib在无血清RPMI1640培养基中处理Jurkat细胞0.5小时,DMSO浓度为0.1%。更换新鲜的含有10%血清的RPMI1640培养基,去除细胞外Talazoparib。然后在不同的时间点收集Jurkat细胞,每个时间点收集1x106细胞,重悬在20μL PBS中,用PCR仪在48.5℃加热三分钟。用含有蛋白酶抑制剂(Roche,瑞士)的细胞裂解液(1%NP-40,150mM NaCl,50mM Tris-HCl pH7.5)冰上裂解细胞半个小时。离心后,收集上清,用SDS-PAGE和免疫印迹检测PARP-1。平行的,用去除细胞外Talazoparib前的Jurkat细胞作为阳性对照,代表了该解离试验的最大起始信号水平和已测定的93%PARP-1占有率(依据已测定的剂量效应曲线)。用0.1%DMSO处理的Jurkat细胞作为阴性对照,代表了该试验条件下的噪音水平。样品中检测到比阴性对照中更高水平的PARP-1,意味着在去除细胞外Talazoparib后该时间点Talazoparib仍与细胞内PARP-1相互作用。用样品中PARP-1水平减去阴性对照中PARP-1水平作为该样品中Talazoparib结合的PARP-1的水平,以时间为函数构建Jurkat细胞内Talazoparib PARP-1相互作用的解离曲线,确定细胞内Talazoparib PARP-1相互作用的解离速率,滞留时间和解离半衰期。依据阳性对照中已知的Talazoparib PARP-1占有率,将细胞内Talazoparib PARP-1相互作用的解离曲线转换为该试验条件下细胞内Talazoparib PARP-1占有率的动力学曲线,确定该试验条件下Talazoparib的治疗持续时间范围。Cell dissociation assay: Jurkat cells were treated with 100 nM Talazoparib in serum-free RPMI 1640 medium for 0.5 hour with a DMSO concentration of 0.1%. Replace the extracellular Talazoparib with fresh RPMI1640 medium containing 10% serum. Then collected at various time points Jurkat cells, 1x10 6 per time point were collected cells were resuspended in 20μL PBS heated at 48.5 deg.] C using a PCR machine for three minutes. The cells were lysed on ice for half an hour with a cell lysate containing protease inhibitor (Roche, Switzerland) (1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5). After centrifugation, the supernatant was collected, and PARP-1 was detected by SDS-PAGE and immunoblotting. Parallel, using Jurkat cells prior to removal of extracellular Talazoparib as a positive control, represents the maximum initial signal level for the dissociation test and the 93% PARP-1 occupancy measured (based on the measured dose effect curve). Jurkat cells treated with 0.1% DMSO as a negative control represent the noise level under the test conditions. A higher level of PARP-1 was detected in the sample than in the negative control, meaning that Talazoparib still interacted with intracellular PARP-1 at this time point after removal of extracellular Talazoparib. The level of PARP-1 in the sample was subtracted from the level of PARP-1 in the negative control as the level of Talazoparib-bound PARP-1 in the sample, and the dissociation curve of the Talazoparib PARP-1 interaction in Jurkat cells was constructed as a function of time to determine the cell. Dissociation rate, retention time and dissociation half-life of the internal Talazoparib PARP-1 interaction. Based on the known Talazoparib PARP-1 occupancy rate in the positive control, the dissociation curve of intracellular Talazoparib PARP-1 interaction was converted to the kinetic curve of intracellular Talazoparib PARP-1 occupancy under the test conditions, and the experimental conditions were determined. The duration of treatment of the lower Talazoparib.
SDS-PAG&免疫印迹:用SDS-PAGE分离PARP-1蛋白,用 Trans-Blot Turbo(Bio-Rad,美国)转印到PVDF膜上,封闭后,用抗PARP-1抗体检测(#sc-8007,SANTA CRUZ BIOTECHNOLOGY,美国),用CCD照相机(GE Healthcare,美国)记录化学发光信号。SDS-PAG & Immunoblotting: Separation of PARP-1 protein by SDS-PAGE Trans-Blot Turbo (Bio-Rad, USA) was transferred onto a PVDF membrane, blocked, and tested with anti-PARP-1 antibody (#sc-8007, SANTA CRUZ BIOTECHNOLOGY, USA) with a CCD camera (GE Healthcare, USA) Record the chemiluminescent signal.
参见图5,该图为Jurkat细胞内PARP-1在缺少(●)和呈现(▲)Talazoparib的情况下的热熔解曲线图;其中,(●)表示未用Talazoparib处理情况下PARP-1的热熔解曲线图;(▲)表示Talazoparib处理情况下PARP-1的热熔解曲线图;其中,纵坐标“相对条带强度”是指实施例2中免疫印迹测定的各数据结果的PARP-1发光强度相对于最强PARP-1发光强度的比值。通过图5所示的热熔解曲线图可知:Talazoparib处理组比对照组在温度区间46-49℃展示了更高水平的PARP-1,所以可选择48.5或49℃作为加热温度用于测定细胞内Talazoparib PARP-1相互作用的持久力。See Figure 5, which is a graph of the thermal melting of PARP-1 in Jurkat cells in the absence (•) and in the presence of (▲) Talazoparib; wherein (●) indicates the heat of PARP-1 in the absence of treatment with Talazoparib Melt curve; (▲) shows the thermal melting curve of PARP-1 in the case of Talazoparib treatment; wherein, the ordinate "relative band intensity" refers to the PARP-1 luminescence intensity of each data result measured by immunoblotting in Example 2. The ratio of the intensity of the strongest PARP-1 luminescence. From the thermal melting curve shown in Figure 5, the Talazoparib treatment group exhibited a higher level of PARP-1 than the control group at a temperature range of 46-49 ° C, so 48.5 or 49 ° C could be selected as the heating temperature for intracellular determination. The persistence of Talazoparib PARP-1 interaction.
参见图6,该图为Jurkat细胞内Talazoparib与PARP-1的剂量效应曲线图。如图6所示,可溶的PARP-1的水平与Talazoparib(0.001-10000nM范围内)展示了浓度依赖的增加,所以可选择100nM Talazoparib,对应细胞内93%PARP-1占有率,作为解离试验中Talazoparib的试验浓度。See Figure 6, which is a graph of the dose response of Talazoparib and PARP-1 in Jurkat cells. As shown in Figure 6, the level of soluble PARP-1 and Talazoparib (in the range of 0.001-10000 nM) showed a concentration-dependent increase, so 100 nM Talazoparib was selected, corresponding to 93% PARP-1 occupancy in cells, as dissociation Test concentration of Talazoparib in the test.
参见图7和图8,图7为Jurkat细胞内Talazoparib与PARP-1相互作用的解离曲线图。图8为Jurka细胞内Talazoparib与PARP-1占有率的动力学曲线图。图7和图8的归一化标准是每个时间点相同数目的细胞。如图7所示,Talazoparib结合的PARP-1的水平随解离时间的增加而非线性降低。应用一个三相解离模型分析Jurkat细胞内Talazoparib与PARP-1相互作用的解离曲线(如图7所示)。第一相是一个快的解离阶段,发生在去除细胞外Talazoparib后第一个小时内,期间约50%Talazoparib PARP-1复合物解离,其解离速率常数为0.69h-1。并且在去除细胞外Talazoparib过程中,没有观察到细胞内Talazoparib PARP-1复合物的解离。第二相是一个非常慢的解离阶段,发生在接下来的23小时内,期间细胞内Talazoparib PARP-1复合物的水平几乎没有变化,其解离速率非常慢难以准确拟合。第三相是一个稍快的解离阶段,细胞内剩余的Talazoparib  PARP-1复合物将逐渐的解离。去除细胞外Talazoparib后36小时仍有约25%Talazoparib PARP-1复合物存在,所以可推测Jurkat细胞内Talazoparib PARP-1相互作用的滞留时间将是超过36小时,远高于已报道的用SPR技术测定的Talazoparib PARP-1滞留时间2.6小时(Clin.Cancer Res.,19,5003-5015,2013)。也可确定细胞内Talazoparib PARP-1解离半衰期为24小时,远高于已报道的用SPR技术测定的Talazoparib PARP-1解离半衰期1.8小时(Clin.Cancer Res.,19,5003-5015,2013)。依据阳性对照中93%PARP-1占有率,将上述Talazoparib细胞内解离曲线(如图7所示)转换为该试验条件下Jurkat细胞内Talazoparib PARP-1占有率的动力学曲线(如图8所示),可从PARP-1占有率动力学曲线中确定在该试验条件下Talazoparib的治疗持续时间范围。例如假设细胞内最低有效的PARP-1占有率为50%,所以在此种情况下Talazoparib的治疗持续时间范围为24小时,意味着清除细胞外Talazoparib后,Talazoparib的细胞活性仍然能够继续维持24小时。Referring to Figures 7 and 8, Figure 7 is a dissociation plot of the interaction of Talazoparib with PARP-1 in Jurkat cells. Figure 8 is a graph showing the kinetics of Talazoparib and PARP-1 occupancy in Jurka cells. The normalization criteria of Figures 7 and 8 are the same number of cells at each time point. As shown in Figure 7, the level of Talazoparib-bound PARP-1 decreased non-linearly with increasing dissociation time. A three-phase dissociation model was used to analyze the dissociation curve of Talazoparib interaction with PARP-1 in Jurkat cells (as shown in Figure 7). The first phase is a fast dissociation phase that occurs during the first hour after removal of extracellular Talazoparib, during which about 50% of the Talazoparib PARP-1 complex dissociates with a dissociation rate constant of 0.69 h -1 . And during the removal of extracellular Talazoparib, no dissociation of intracellular Talazoparib PARP-1 complex was observed. The second phase is a very slow dissociation phase that occurs during the next 23 hours during which there is little change in the level of intracellular Talazoparib PARP-1 complex, and its dissociation rate is very slow and difficult to fit accurately. The third phase is a slightly faster dissociation phase, and the remaining Talazoparib PARP-1 complex in the cell will gradually dissociate. About 25% of the Talazoparib PARP-1 complex is still present 36 hours after removal of extracellular Talazoparib, so it is speculated that the retention time of Talazoparib PARP-1 interaction in Jurkat cells will be more than 36 hours, much higher than the reported SPR technique. The measured Talazoparib PARP-1 retention time was 2.6 hours (Clin. Cancer Res., 19, 5003-5015, 2013). It was also determined that the intracellular Talazoparib PARP-1 dissociation half-life was 24 hours, which was much higher than the reported Talazoparib PARP-1 dissociation half-life determined by the SPR technique for 1.8 hours (Clin. Cancer Res., 19, 5003-5015, 2013). ). Based on the 93% PARP-1 occupancy rate in the positive control, the intracellular dissociation curve of Talazoparib (shown in Figure 7) was converted to the kinetic curve of Talazoparib PARP-1 occupancy in Jurkat cells under the experimental conditions (Fig. 8). As shown, the duration of treatment of Talazoparib under the test conditions can be determined from the PARP-1 occupancy kinetics curve. For example, assuming that the lowest effective PARP-1 occupancy in the cell is 50%, in this case the treatment duration of Talazoparib is in the range of 24 hours, meaning that the cell activity of Talazoparib can continue to be maintained for 24 hours after the removal of extracellular Talazoparib. .
此外,比较实施例1和实施例2可发现Jurkat细胞内Talazoparib PARP-1相互作用比星形孢菌素Aurora A相互作用具有更强的结合持久力,意味着同一细胞内不同的配体与靶蛋白组合可产生不同的结合持久力。Furthermore, in Comparative Example 1 and Example 2, it was found that the Talazoparib PARP-1 interaction in Jurkat cells has stronger binding endurance than the staurosporin Aurora A interaction, meaning different ligands and targets in the same cell. Protein combinations can produce different binding endurance.
实施例3Example 3
本实施例的目的是证明细胞增殖可减弱细胞内配体靶蛋白相互作用的持久力以及细胞内药理作用的持续时间。为了验证细胞增殖的影响,本试验采用固定细胞取样体积作为归一化的标准,模拟细胞增殖速度为0的情况,在此种情况下,每个时间点样品中可能包含不同数目的细胞,但拥有相同起始水平的配体靶蛋白复合物。The purpose of this example was to demonstrate that cell proliferation can attenuate the persistence of intracellular ligand target protein interactions and the duration of intracellular pharmacological effects. In order to verify the effects of cell proliferation, this experiment uses a fixed cell sampling volume as a normalization standard to simulate a cell proliferation rate of 0. In this case, the sample may contain a different number of cells at each time point, but Ligand target protein complexes with the same starting level.
用50nM Talazoparib无血清RPMI1640培养基中处理Jurkat细胞0.5小时,DMSO浓度为0.1%。更换新鲜的含有10%血清的RPMI1640培养基,以此去除细胞外Talazoparib。然后每个检测时间点收集1ml细胞,计数后,重悬在20μL PBS中,用PCR仪48.5℃加热三分钟。用含有 蛋白酶抑制剂(Roche,瑞士)的细胞裂解液(1%NP-40,150mM NaCl,50mM Tris-HCl pH7.5)冰上裂解半个小时。离心后,收集上清,用SDS-PAGE和免疫印迹检测PARP-1。平行的,用去除细胞外Talazoparib前的细胞作为阳性对照,代表了该解离试验的最大起始信号水平和已计算的85%PARP-1占有率(依据以上剂量效应曲线)。用0.1%DMSO处理的1x106细胞作为阴性对照,代表了该情况下的噪音水平。用样品中PARP-1的水平减去阴性对照中PARP-1的水平乘以增殖率的背景噪音,作为该样品中Talazoparib结合的PARP-1的水平,以解离时间为函数构建在此试验条件下Jurkat细胞内Talazoparib PARP-1相互作用的解离曲线,确定细胞内Talazoparib PARP-1相互作用的解离速率,滞留时间和解离半衰期。依据阳性对照中已知的Talazoparib PARP-1占有率,将细胞内Talazoparib PARP-1相互作用的解离曲线转换为该试验条件下细胞内Talazoparib PARP-1占有率的动力学曲线,确定该试验条件下Talazoparib的治疗持续时间范围。Jurkat cells were treated with 50 nM Talazoparib serum-free RPMI 1640 medium for 0.5 hour with a DMSO concentration of 0.1%. Extracellular Talazoparib was removed by replacing fresh RPMI1640 medium containing 10% serum. Then, 1 ml of cells were collected at each detection time point, counted, resuspended in 20 μL of PBS, and heated by a PCR instrument at 48.5 ° C for three minutes. Cell lysate (1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 7.5) containing protease inhibitor (Roche, Switzerland) was lysed on ice for half an hour. After centrifugation, the supernatant was collected, and PARP-1 was detected by SDS-PAGE and immunoblotting. Parallel, using cells prior to removal of extracellular Talazoparib as a positive control, represents the maximum initial signal level for the dissociation test and the calculated 85% PARP-1 occupancy (based on the above dose-response curve). 1x10 6 cells treated with 0.1% DMSO served as a negative control, representing the noise level in this case. The level of PARP-1 in the sample was subtracted from the level of PARP-1 in the negative control multiplied by the background noise of the proliferation rate as the level of Talazoparib-bound PARP-1 in the sample, constructed as a function of the dissociation time. The dissociation curve of the Talazoparib PARP-1 interaction in the Jurkat cells determines the dissociation rate, retention time and dissociation half-life of the intracellular Talazoparib PARP-1 interaction. Based on the known Talazoparib PARP-1 occupancy rate in the positive control, the dissociation curve of intracellular Talazoparib PARP-1 interaction was converted to the kinetic curve of intracellular Talazoparib PARP-1 occupancy under the test conditions, and the experimental conditions were determined. The duration of treatment of the lower Talazoparib.
用SDS-PAGE分离PARP-1蛋白,用Trans-Blot Turbo(Bio-Rad,美国)转印到PVDF膜上。封闭后,用抗PARP-1抗体检测(#sc-8007,SANTA CRUZ BIOTECHNOLOGY,美国),用CCD照相机(GE Healthcare,美国)记录化学发光信号。The PARP-1 protein was separated by SDS-PAGE and transferred to a PVDF membrane using Trans-Blot Turbo (Bio-Rad, USA). After blocking, the chemiluminescent signal was recorded with a CCD camera (GE Healthcare, USA) using an anti-PARP-1 antibody assay (#sc-8007, SANTA CRUZ BIOTECHNOLOGY, USA).
参见图9和图10,图9为Jurkat细胞内Talazoparib与PARP-1相互作用的解离曲线图,归一化标准是每个时间点相同体积的细胞,模拟细胞增速度为0的情况。图10为Jurkat细胞内Talazoparib与PARP-1占有率的动力学曲线图,归一化标准是每个时间点相同体积的细胞,模拟细胞增速度为0的情况。如图9所示的解离曲线图,Talazoparib结合的PARP-1的水平随解离时间的增加而非线性降低。应用一个两相解离模型分析本实施例试验条件下Jurkat细胞内Talazoparib与PARP-1相互作用的解离曲线图。相似的,第一相是一个快的解离阶段,发生在去除细胞外Talazoparib后第一个小时内,期间约50%Talazoparib PARP-1复合物解离,其解离速率常数为0.62h-1。再次确认在去除细胞外Talazoparib过程中,没有观察 到细胞内Talazoparib PARP-1复合物的解离。第二相是一个非常慢的解离阶段,发生在接下来的35小时内,期间细胞内Talazoparib PARP-1复合物的水平几乎没有变化,其解离速率非常慢,难以准确拟合。尤其是36小时时间点仍然有约60%Talazoparib PARP-1复合物存在,所以在此试验条件下Jurkat细胞内Talazoparib与PARP-1相互作用的滞留时间是远多于36小时,细胞内Talazoparib与PARP-1解离半衰期也多于36小时。依据阳性对照中85%PARP-1占有率,将上述Talazoparib细胞内解离曲线(如图9所示)转换为该试验条件下Jurkat细胞内Talazoparib PARP-1占有率的动力学曲线(如图10所示),可从PARP-1占有率动力学曲线中确定在此种情况下Talazoparib的治疗持续时间范围。相似的假设细胞内最低有效的PARP-1占有率为50%,所以该试验条件下Talazoparib治疗持续时间范围为多于36小时,意味着清除细胞外Talazoparib后,Talazoparib的细胞活性仍然能够继续维持36小时。Referring to Figures 9 and 10, Figure 9 is a dissociation plot of Talazoparib interaction with PARP-1 in Jurkat cells. The normalization criterion is the same volume of cells at each time point, simulating a cell growth rate of zero. Figure 10 is a graph showing the kinetics of Talazoparib and PARP-1 occupancy in Jurkat cells. The normalization standard is the same volume of cells at each time point, and the simulated cell growth rate is zero. As shown in the dissociation plot of Figure 9, the level of Talazoparib-bound PARP-1 decreases non-linearly with increasing dissociation time. A dissociation curve of the interaction between Talazoparib and PARP-1 in Jurkat cells under the experimental conditions of this example was analyzed using a two-phase dissociation model. Similarly, the first phase is a fast dissociation phase that occurs during the first hour after removal of extracellular Talazoparib, during which about 50% of the Talazoparib PARP-1 complex dissociates with a dissociation rate constant of 0.62 h -1 . It was confirmed again that no dissociation of the intracellular Talazoparib PARP-1 complex was observed during the removal of extracellular Talazoparib. The second phase is a very slow dissociation phase that occurs during the next 35 hours during which there is little change in the level of intracellular Talazoparib PARP-1 complex, and the dissociation rate is very slow and difficult to fit accurately. In particular, about 60% of the Talazoparib PARP-1 complex was still present at the 36-hour time point, so the retention time of Talazoparib interacting with PARP-1 in Jurkat cells was much longer than 36 hours under this test condition. Intracellular Talazoparib and PARP The -1 dissociation half-life is also more than 36 hours. Based on the 85% PARP-1 occupancy rate in the positive control, the intracellular dissociation curve of Talazoparib (shown in Figure 9) was converted to the kinetic curve of Talazoparib PARP-1 occupancy in Jurkat cells under the experimental conditions (Fig. 10). As shown), the duration of treatment of Talazoparib in this case can be determined from the PARP-1 occupancy kinetic curve. A similar hypothesis is that the least potent PARP-1 occupancy rate in the cell is 50%, so the duration of Talazoparib treatment is more than 36 hours under this test condition, meaning that the cell viability of Talazoparib can continue to be maintained after removal of extracellular Talazoparib. hour.
实施例2采用相同细胞数目的归一化方法,代表了正常的细胞增殖速度,实施例3采用固定细胞取样体积的归一化方法,模拟了细胞增殖速度为0的情况。比较两种情况下的Talazoparib PARP-1的解离曲线(如图7和9所示),尤其是在36小时时间点,以及解离半衰期,可发现实施例3比实施例2具有更强的细胞内Talazoparib PARP-1相互作用的持久力,证明细胞增殖可以减弱细胞内配体靶蛋白相互作用的持久力。比较两种情况下的Talazoparib PARP-1占有率的动力学曲线(如图8和10所示)和治疗持续时间范围,可发现实施例3即使应用了稍低的Talazoparib试验浓度仍比实施例2具有更长的治疗持续时间范围,证明细胞增殖可以减弱细胞药理作用的持续时间。Example 2 uses the normalization method of the same cell number to represent the normal cell proliferation rate, and Example 3 uses the normalization method of the fixed cell sampling volume to simulate the case where the cell proliferation rate is zero. Comparing the dissociation curves of Talazoparib PARP-1 in both cases (as shown in Figures 7 and 9), especially at the 36 hour time point, and the dissociation half-life, Example 3 was found to be stronger than Example 2. The persistence of intracellular Talazoparib PARP-1 interactions demonstrates that cell proliferation can attenuate the persistence of ligand-target protein interactions in cells. Comparing the kinetic curves of Talazoparib PARP-1 occupancy (as shown in Figures 8 and 10) and the duration of treatment in both cases, it can be found that Example 3 is still more effective than Example 2 even if a slightly lower Talazoparib test concentration is applied. With a longer duration of treatment, it is demonstrated that cell proliferation can attenuate the duration of cellular pharmacological effects.
上述仅为本发明的部分优选实施例,本发明并不仅限于实施例的内容。对于本领域中的技术人员来说,在本发明技术方案的构思范围内可以有各种变化和更改,所作的任何变化和更改,均在本发明保护范围之内。 The above is only some of the preferred embodiments of the present invention, and the present invention is not limited to the contents of the embodiments. It will be apparent to those skilled in the art that various changes and modifications may be made without departing from the scope of the invention.

Claims (13)

  1. 一种测定细胞内配体靶蛋白相互作用持久力的方法,该方法包括如下步骤:A method for determining the endurance of a ligand protein interaction in a cell, the method comprising the steps of:
    步骤1,用所述配体处理所述细胞;Step 1, treating the cells with the ligand;
    步骤2,去除所述细胞外的所述配体;Step 2, removing the ligand outside the extracellular;
    步骤3,收集去除所述配体后不同时间点的所述细胞;Step 3, collecting the cells at different time points after removing the ligand;
    步骤4,加热所述细胞;Step 4, heating the cells;
    步骤5,裂解所述细胞;Step 5, lysing the cells;
    步骤6,分析细胞裂解液中可溶的或不可溶的所述靶蛋白的水平随时间变化的函数。In step 6, a function of the level of soluble or insoluble target protein in the cell lysate as a function of time is analyzed.
  2. 如权利要求1所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:所述配体是细胞代谢物,小分子或药物;所述靶蛋白是细胞内与配体相互作用的蛋白分子;所述细胞是哺乳动物细胞,细胞株,工程细胞或原代细胞。The method for determining the endurance of an intracellular ligand target protein interaction according to claim 1, wherein the ligand is a cell metabolite, a small molecule or a drug; and the target protein is an intracellular and ligand interaction with each other. A protein molecule that acts; the cell is a mammalian cell, a cell strain, an engineered cell, or a primary cell.
  3. 如权利要求1所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:所述步骤1中将配体加入到细胞培养基中,进入细胞后与靶蛋白相互作用。The method for determining the endurance of an intracellular ligand target protein interaction according to claim 1, wherein in the step 1, the ligand is added to the cell culture medium and enters the cell to interact with the target protein.
  4. 如权利要求1所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:所述步骤2中去除细胞外配体的方法包括更换新鲜的培养基或用溶液清洗细胞后更换新鲜培养基。The method for determining the endurance of an intracellular ligand target protein interaction according to claim 1, wherein the method for removing the extracellular ligand in the step 2 comprises replacing the fresh medium or replacing the cells with the solution and then replacing the cells. Fresh medium.
  5. 如权利要求1所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:所述步骤3中收集去除细胞外配体后一个或多个时间点的细胞样品。The method for determining the endurance of an intracellular ligand target protein interaction according to claim 1, wherein said step 3 collects a cell sample at one or more time points after removal of the extracellular ligand.
  6. 如权利要求1所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:所述步骤4中加热细胞到一个配体结合的和非结合的靶蛋白有不同热稳定性的温度。The method for determining the endurance of an intracellular ligand target protein interaction according to claim 1, wherein the step of heating the cells to a ligand-bound and unbound target protein has different thermal stability. temperature.
  7. 如权利要求1所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特 征在于:所述步骤5中裂解所述细胞后,还包括提取细胞内蛋白,用离心或过滤的方法分离可溶的和不可溶的细胞裂解液组分。A method for determining the endurance of interaction of a target ligand protein in a cell according to claim 1, wherein The method further comprises: after lysing the cells in the step 5, further comprising extracting intracellular proteins, and separating the soluble and insoluble cell lysate fractions by centrifugation or filtration.
  8. 如权利要求1所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:所述步骤6中分析靶蛋白的水平利用靶蛋白定量进行确定,靶蛋白的定量方法包括抗体技术,质谱法,靶蛋白的生物活性或融合标签的酶活性。The method for determining the endurance of an intracellular ligand target protein interaction according to claim 1, wherein the level of the target protein in the step 6 is determined by quantitative use of a target protein, and the quantitative method of the target protein includes antibody technology. , mass spectrometry, biological activity of the target protein or enzymatic activity of the fusion tag.
  9. 如权利要求1所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:所述步骤6具体包括测量可溶的靶蛋白,确定细胞内配体结合的靶蛋白的水平,以解离时间为函数构建细胞内配体靶蛋白相互作用的解离曲线,计算细胞内配体靶蛋白相互作用的解离速率常数,滞留时间和解离半衰期。The method for determining the endurance of an intracellular ligand target protein interaction according to claim 1, wherein the step 6 specifically comprises measuring a soluble target protein and determining the level of a target protein bound by the ligand in the cell, The dissociation curve of intracellular ligand target protein interaction was constructed by dissociation time, and the dissociation rate constant, retention time and dissociation half-life of intracellular ligand target protein interaction were calculated.
  10. 如权利要求1所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:还包括收集去除细胞外配体前的细胞样品。The method for determining the endurance of an intracellular ligand target protein interaction according to claim 1, further comprising collecting a cell sample before removing the extracellular ligand.
  11. 如权利要求10所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:细胞内配体靶蛋白相互作用的解离反应包括去除细胞外配体期间的解离反应和去除细胞外配体后的解离反应。The method for determining the endurance of an intracellular ligand target protein interaction according to claim 10, wherein the dissociation reaction of the intracellular ligand target protein interaction comprises dissociation reaction and removal during removal of the extracellular ligand. Dissociation reaction after extracellular ligand.
  12. 如权利要求1所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:还包括配体处理后但未去除的阳性对照,有已知的靶蛋白占有率水平。A method of determining the persistence of interaction of a target ligand protein in an intracellular ligand according to claim 1, further comprising a positive control after ligand treatment but not removed, having a known target protein occupancy level.
  13. 如权利要求12所述的测定细胞内配体靶蛋白相互作用持久力的方法,其特征在于:确定去除细胞外配体后不同时间点的靶蛋白占有率,构建细胞内配体靶蛋白占有率的动力学曲线,计算高于最低有效靶蛋白占有率的配体的治疗持续时间范围。 The method for determining the endurance of interaction of a target ligand protein in an intracellular ligand according to claim 12, wherein the target protein occupancy rate at different time points after removal of the extracellular ligand is determined, and the occupancy rate of the ligand protein in the cell is constructed. The kinetic curve calculates the duration of treatment of the ligand above the lowest effective target protein occupancy.
PCT/CN2016/083998 2016-05-31 2016-05-31 Method for determining endurance of interaction between ligand and target protein in cell WO2017206041A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201680003155.8A CN109690316A (en) 2016-05-31 2016-05-31 The method for measuring intracellular ligand target protein interaction endurance
PCT/CN2016/083998 WO2017206041A1 (en) 2016-05-31 2016-05-31 Method for determining endurance of interaction between ligand and target protein in cell

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2016/083998 WO2017206041A1 (en) 2016-05-31 2016-05-31 Method for determining endurance of interaction between ligand and target protein in cell

Publications (1)

Publication Number Publication Date
WO2017206041A1 true WO2017206041A1 (en) 2017-12-07

Family

ID=60479401

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/083998 WO2017206041A1 (en) 2016-05-31 2016-05-31 Method for determining endurance of interaction between ligand and target protein in cell

Country Status (2)

Country Link
CN (1) CN109690316A (en)
WO (1) WO2017206041A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112924693B (en) * 2019-12-06 2022-09-27 中国科学院大连化学物理研究所 Protein thermal stability analysis method for synergistic quantification of supernatant and precipitate

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101223447A (en) * 2005-06-14 2008-07-16 塞尔卓姆股份公司 Process for the identification of novel enzyme interacting compounds
CN103645324A (en) * 2013-12-03 2014-03-19 胡朝阳 High-throughout drug target screening method based on cell heat transferring technology
CN103733067A (en) * 2011-04-18 2014-04-16 艾维特拉普罗特玛公司 Methods for determining ligand binding to a target protein using a thermal shift assay
WO2014093677A1 (en) * 2012-12-12 2014-06-19 Promega Corporation Recognition of cellular target binding by a bioactive agent using intracellular bioluminescence resonance energy transfer

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104897826B (en) * 2015-06-16 2016-06-22 广西师范大学 A kind of method detecting micromolecular compound and target protein interaction for cell in-situ

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101223447A (en) * 2005-06-14 2008-07-16 塞尔卓姆股份公司 Process for the identification of novel enzyme interacting compounds
CN103733067A (en) * 2011-04-18 2014-04-16 艾维特拉普罗特玛公司 Methods for determining ligand binding to a target protein using a thermal shift assay
WO2014093677A1 (en) * 2012-12-12 2014-06-19 Promega Corporation Recognition of cellular target binding by a bioactive agent using intracellular bioluminescence resonance energy transfer
CN103645324A (en) * 2013-12-03 2014-03-19 胡朝阳 High-throughout drug target screening method based on cell heat transferring technology

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
COPELAND, R.A. ET AL.: "Drug-target residence time and its implications for lead optimization", NATURE REVIEWS DRUG DISCOVERY, vol. 5, 1 September 2006 (2006-09-01), pages 730 - 739, XP002570141, Retrieved from the Internet <URL:doi:10.1038/nrd2082> *
JAFARI, R. ET AL.: "The cellular thermal shift assay for evaluating drug target interactions in cells", NATURE PROTOCOLS, vol. 9, no. 9, 7 August 2014 (2014-08-07), pages 2100 - 2122, XP055193929, Retrieved from the Internet <URL:doi:10.1038/nprot.2014.138> *
MOREAU, MORGANE J.J. ET AL.: "Quantitative determination of protein stability and ligand binding using a green fluorescent protein reporter system", MOL. BIOSYST., vol. 6, 7 May 2010 (2010-05-07), pages 1285 - 1292, XP055027468, Retrieved from the Internet <URL:doi:10.1039/c002001j> *
REDHEAD, M. ET AL.: "A combinatorial biophysical approach; FTSAand SPR for identifying small molecule ligands and PAINs", ANALYTICAL BIOCHEMISTRY, vol. 479, 30 March 2015 (2015-03-30), pages 63 - 73, XP029157506, Retrieved from the Internet <URL:doi:10.1016/j.ab.2015.03.013> *

Also Published As

Publication number Publication date
CN109690316A (en) 2019-04-26

Similar Documents

Publication Publication Date Title
Miettinen et al. Thermal proteome profiling of breast cancer cells reveals proteasomal activation by CDK 4/6 inhibitor palbociclib
Friman Mass spectrometry-based Cellular Thermal Shift Assay (CETSA®) for target deconvolution in phenotypic drug discovery
Wang et al. Mapping sites of aspirin-induced acetylations in live cells by quantitative acid-cleavable activity-based protein profiling (QA-ABPP)
DK2699910T3 (en) A method for determining ligand binding to a target protein using a thermal shift assay
JP5364096B2 (en) Method for detecting substance interaction
He et al. Treatment of hypertension by increasing impaired endothelial TRPV 4‐KC a2. 3 interaction
WO2010141596A2 (en) Chemical proteomic assay for optimizing drug binding to target proteins
Daniels et al. Examining the complexity of human RNA polymerase complexes using HaloTag technology coupled to label free quantitative proteomics
Roehrl et al. Discovery of small-molecule inhibitors of the NFAT− calcineurin interaction by competitive high-throughput fluorescence polarization screening
Huang et al. NMR-based investigation into protein phosphorylation
Busby et al. Advancements in assay technologies and strategies to enable drug discovery
Valtonen et al. Nanomolar protein–protein interaction monitoring with a label-free Protein-Probe technique
Somsen et al. Reversible dual-covalent molecular locking of the 14-3-3/ERRγ protein–protein interaction as a molecular glue drug discovery approach
Langlois et al. A GID E3 ligase assembly ubiquitinates an Rsp5 E3 adaptor and regulates plasma membrane transporters
Wang et al. Optimized data-independent acquisition approach for proteomic analysis at single-cell level
Vamisetti et al. Selective macrocyclic peptide modulators of Lys63-linked ubiquitin chains disrupt DNA damage repair
Henneberg et al. Activity-based profiling of cullin–RING E3 networks by conformation-specific probes
Gao et al. Chemoproteomics, A Broad Avenue to Target Deconvolution
WO2017206041A1 (en) Method for determining endurance of interaction between ligand and target protein in cell
Gally et al. Identifying bioactivity of pseudo-natural products using the Cell Painting assay.
Mok et al. The in vivo dynamic interplay of MDC1 and 53BP1 at DNA damage-induced nuclear foci
Schwalm et al. Targeting LC3/GABARAP for degrader development and autophagy modulation
Sheahan et al. A molecular imaging biosensor detects in vivo protein folding and misfolding
Ciulli et al. PROTAC degraders: Mechanism, recent advances, and future challenges
Man et al. Super-resolution imaging of autophagy by a preferred pair of self-labeling protein tags and fluorescent ligands

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16903429

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205N DATED 09/04/2019)

122 Ep: pct application non-entry in european phase

Ref document number: 16903429

Country of ref document: EP

Kind code of ref document: A1