We provide robust and sensitive in vitro screening and characterization platforms for accelerating the discovery and screening of potential therapies for hemophilia. Our services are designed to evaluate the activity, binding, and efficacy of candidate drugs targeting coagulation factors, inhibitors, and related pathways implicated in hemophilia. Key targets include Factor VIII, Factor IX, and their regulatory proteins, as well as pathways governing coagulation and fibrinolysis. Our assays enable assessment of coagulation activity, inhibitor potency, and the molecular mechanisms underlying the bleeding disorder.
We offer a comprehensive suite of biochemical and cell-based in vitro assays to characterize drug candidates for hemophilia. These methods assess molecular binding, enzymatic activity, expression, and cellular responses, allowing for multifaceted analysis of therapeutic efficacy and mechanism of action. The overall purpose is to provide robust data supporting lead optimization and preclinical decision-making.
Biolayer interferometry assay: Measures real-time biomolecular interactions, enabling evaluation of binding kinetics and affinity between therapeutic candidates and target proteins relevant to hemophilia.
D-Ile-Pro-Arg-p-nitroanilide as substrate: Utilized for enzymatic assays to quantify the activity of coagulation factors or inhibitors by monitoring substrate cleavage.
Displacement of [3H]-vasopressin: Assesses binding competition and receptor interactions, offering insights into drug-target specificity.
ELISA assay: Provides quantitative detection of proteins, antibodies, or inhibitors in biological samples, supporting pharmacodynamic and pharmacokinetic studies.
Flow cytometry assay: Enables detailed analysis of cellular markers, activation status, and intracellular signaling in blood or cell-based models.
Fluorescent assay: Detects enzyme activity or molecular interactions using fluorescence-based readouts for sensitive and high-throughput screening.
Gene reporter assay: Monitors gene expression changes in response to drug treatments, elucidating effects on coagulation-related pathways.
RNA assay: Quantifies mRNA levels of coagulation factors or pathway components to assess gene regulation by therapeutic agents.
Radioactivity assay: Utilizes radiolabeled substrates or ligands to sensitively measure enzymatic activity, binding, or metabolic fate.
Surface plasmon resonance assay: Provides detailed kinetic and affinity data on drug-target interactions, supporting lead optimization.
Our assays measure key pharmacological parameters such as potency, binding affinity, and minimal effective dose, which are essential for characterizing and comparing candidate therapeutics. These quantitative metrics inform structure-activity relationships and guide the selection of promising compounds for further development. Understanding these parameters is critical for optimizing efficacy and safety profiles.
EC-50: The concentration of a drug that produces 50% of its maximal effect, indicating potency and guiding dose selection.
IC-50: The concentration of an inhibitor that reduces a specific biological activity by 50%, used to compare inhibitor strengths.
Kd: The equilibrium dissociation constant reflecting the binding affinity between a drug and its target; lower values indicate stronger binding.
Ki: The inhibition constant representing a compound's potency in inhibiting a specific target, important for ranking inhibitors.
MED: The minimal effective dose required to elicit a detectable biological response, useful for defining therapeutic windows.
Apolipoprotein B (ApoB) is investigated in hemophilia drug development due to its involvement in lipid metabolism and potential impact on coagulation pathways. ApoB testing, using sensitive RNA assays, helps evaluate drug effects on ApoB expression. Measuring the IC-50 parameter enables quantification of drug potency in inhibiting ApoB-related targets, supporting the development of safer, more effective hemophilia therapies.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Gene (APOB) transcription, inhibition | Hepatocytes (primary), human | RNA assay | IC-50 |
| Gene (APOB) transcription, inhibition | Hepatocytes (primary), mouse | RNA assay | IC-50 |
The Arginine Vasopressin Receptor 2 (AVPR2) modulates water reabsorption and can influence bleeding risk in hemophilia. AVPR2 testing is vital for evaluating potential hemophilia therapies targeting this pathway. Using gene reporter assays, fluorescent assays, and [3H]-vasopressin displacement, we measure key parameters—Ki (inhibitory constant), MED (minimum effective dose), and EC-50 (half-maximal effective concentration)—to assess drug efficacy and receptor interaction.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Gene transcription, induction | HEK293 human embryonic kidney cells transfected with human V2 receptor | Gene reporter assay | EC-50 |
| Urine volume increase, inhibition | Bladder, rat | MED | |
| Vasopressin V2 receptor affinity | CHO Chinese hamster ovary cells transfected with human V2 receptor | Displacement of [3H]-vasopressin | Ki |
| Vasopressin V2 receptor affinity | CHO Chinese hamster ovary cells transfected with human receptor | Displacement of [3H]-vasopressin | Ki |
| Vasopressin V2 receptor affinity | HeLa human cervix adenocarcinoma cells transfected with human receptor | Displacement of [3H]-vasopressin | Ki |
| Vasopressin V2 receptor affinity | Kidney (medulla), dog | Displacement of [3H]-vasopressin | Ki |
| Vasopressin V2 receptor affinity | Kidney (medulla), rabbit | Displacement of [3H]-vasopressin | Ki |
| Vasopressin V2 receptor affinity | Kidney, monkey | Displacement of [3H]-vasopressin | Ki |
| Vasopressin V2 receptor affinity | Kidney, mouse | Displacement of [3H]-vasopressin | Ki |
| Vasopressin V2 receptor affinity | Kidney, rat | Displacement of [3H]-vasopressin | Ki |
| Vasopressin V2 receptor affinity | LV2 mouse fibroblasts transfected with human receptor | Displacement of [3H]-vasopressin | Ki |
| cAMP production, induction | CHO Chinese hamster ovary cells transfected with human V2 receptor | EC-50 | |
| cAMP production, induction | Cells transfected with human V2 receptor | Fluorescent assay | EC-50 |
| cAMP production, induction | HeLa human cervix adenocarcinoma cells transfected with human V2 receptor | EC-50 | |
| cAMP production, induction | LV2 mouse fibroblasts transfected with human V2 receptor | EC-50 |
Coagulation Factor II, Thrombin, plays a central role in hemophilia by converting fibrinogen to fibrin and amplifying coagulation. Thrombin testing is essential in hemophilia drug development to assess therapeutic efficacy and safety. Key methods include chromogenic and clot-based assays, with main parameters such as thrombin generation, activity, and inhibition. Accurate measurement ensures optimal dosing and monitoring of novel hemophilia treatments.
| Pharmacological Activity | Parameter |
|---|---|
| Thrombin, inhibition | IC-50 |
Coagulation Factor IX is essential for proper blood clotting, and its deficiency causes hemophilia B. Accurate Factor IX testing is crucial for hemophilia drug development to assess drug efficacy and safety. Our service employs ELISA, surface plasmon resonance, and biolayer interferometry assays to evaluate interactions and inhibition. Key parameters measured include Kd (binding affinity) and IC50 (inhibitory concentration), ensuring comprehensive characterization of candidate therapeutics.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Coagulation factor IX affinity | Cynomolgus monkey factor | Surface plasmon resonance assay | Kd |
| Coagulation factor IX affinity | Human factor | Surface plasmon resonance assay | Kd |
| Coagulation factor IX affinity | Recombinant human factor | Biolayer interferometry assay | Kd |
| Coagulation factor IX affinity | Recombinant human factor | ELISA assay | IC-50 |
| Coagulation factor IX, inhibition | HEK293 human embryonic kidney cells transfected with mutant factor | ELISA assay | IC-50 |
| Coagulation factor IX, inhibition | Plasma, human | IC-50 | |
| Coagulation factor IXa affinity | Human factor | Surface plasmon resonance assay | Kd |
| Coagulation factor IXa affinity | Kd |
Coagulation Factor VII plays a crucial role in initiating the coagulation cascade, making its assessment vital in hemophilia drug development. Our testing service utilizes D-Ile-Pro-Arg-p-nitroanilide as a chromogenic substrate and biolayer interferometry assays to evaluate drug interactions. Key parameters measured include the dissociation constant (Kd) and half-maximal inhibitory concentration (IC-50), providing critical data on drug efficacy and binding affinity.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Coagulation factor VIIa affinity | Human factor | Biolayer interferometry assay | Kd |
| Coagulation factor VIIa/tissue factor complex, inhibition | Recombinant human factor | D-Ile-Pro-Arg-p-nitroanilide as substrate | IC-50 |
Coagulation Factor VIII is essential for blood clotting, and its deficiency causes hemophilia A. Factor VIII testing is crucial in hemophilia drug development to assess therapeutic efficacy and safety. Key methods include chromogenic and one-stage clotting assays. Main parameters measured are Factor VIII activity levels, recovery, and inhibitor formation, providing vital data to guide dosing, monitor response, and ensure the safety of new treatments.
| Pharmacological Activity | Method | Parameter |
|---|---|---|
| Coagulation Factor VIII affinity | Displacement of von Willebrand factor | IC-50 |
Coagulation Factor X is essential in the blood clotting cascade, and its assessment is vital for hemophilia drug development. Our testing service utilizes surface plasmon resonance assays to evaluate drug interactions with Factor X, providing key parameters such as dissociation constant (Kd) and half-maximal inhibitory concentration (IC-50). These measurements are critical for determining drug potency and binding affinity, ensuring the development of effective hemophilia therapeutics.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Coagulation factor X affinity | Cynomolgus monkey factor | Surface plasmon resonance assay | Kd |
| Coagulation factor X affinity | Human factor | Surface plasmon resonance assay | Kd |
| Coagulation factor X affinity | Kd | ||
| Coagulation factor Xa affinity | Human factor | Surface plasmon resonance assay | Kd |
| Coagulation factor Xa, inhibition | IC-50 |
Protein C, an inactivator of coagulation factors Va and VIIIa, plays a pivotal role in regulating blood clotting, making its assessment crucial in hemophilia drug development. Our testing service utilizes ELISA, surface plasmon resonance, and biolayer interferometry assays to quantify Protein C activity and its interactions. Key parameters measured include Kd (binding affinity) and IC-50 (inhibitory concentration), providing essential data for evaluating therapeutic efficacy and safety.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Protein C (activated) affinity | Human protein | Biolayer interferometry assay | Kd |
| Protein C (activated) affinity | Human protein | ELISA assay | IC-50 |
| Protein C (activated) affinity | Human protein | Surface plasmon resonance assay | Kd |
| Protein C (activated) affinity | Human protein (Phe-Pro-Arg-chloromethylketone active-site-blocked) | Surface plasmon resonance assay | Kd |
| Protein C (activated) affinity | Purified dog protein | ELISA assay | IC-50 |
| Protein C (activated) affinity | Purified human protein | ELISA assay | IC-50 |
| Protein C (activated) affinity | Purified human protein | Surface plasmon resonance assay | Kd |
| Protein C (activated) affinity | Purified monkey protein | ELISA assay | IC-50 |
| Protein C (activated) affinity | Purified rabbit protein | ELISA assay | IC-50 |
| Protein C (activated), inhibition | IC-50 | ||
| Protein C affinity | Human protein | ELISA assay | IC-50 |
Serpin Family C Member 1 (antithrombin) regulates blood coagulation and is critical in hemophilia drug development. Testing its interaction with drug candidates ensures efficacy and safety. Using surface plasmon resonance assays, we determine key parameters such as minimum effective dose (MED) and binding affinity (Kd), providing precise data to optimize therapeutic design and dosing for hemophilia treatments.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Antithrombin affinity | Human protein | Surface plasmon resonance assay | Kd |
| Antithrombin affinity | Human protein | Kd | |
| Antithrombin, inhibition | Plasma, mouse | MED |
Serpin Family E Member 2 (SERPINE2) modulates fibrinolysis and can influence bleeding risk in hemophilia. Testing its activity is crucial for evaluating drug candidates targeting coagulation pathways. Using biolayer interferometry assays, SERPINE2 inhibition or binding by compounds is measured, with IC-50 values quantifying inhibitor potency. This service supports hemophilia drug development by providing precise, real-time assessment of SERPINE2 modulation.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Glia-derived nexin (GDN; SERPINE2) affinity | Recombinant human enzyme | Biolayer interferometry assay | IC-50 |
| Glia-derived nexin (GDN; SERPINE2) affinity | Recombinant mouse enzyme | Biolayer interferometry assay | IC-50 |
Tissue Factor Pathway Inhibitor (TFPI) regulates coagulation and is a key target in hemophilia drug development. TFPI testing is crucial to assess drug efficacy and safety. We offer ELISA, surface plasmon resonance, flow cytometry, and radioactivity assays to evaluate TFPI interactions and inhibition. Main parameters measured include dissociation constant (Kd), half-maximal inhibitory concentration (IC-50), and half-maximal effective concentration (EC-50), providing comprehensive insights for candidate evaluation.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Coagulation factor Xa activation (TFPI-depressed), induction | Human factor | ELISA assay | EC-50 |
| Coagulation factor Xa activation (TFPI-depressed), induction | EC-50 | ||
| Coagulation factor Xa activation (coagulation factor VIIa/tissue factor-depressed), induction | EC-50 | ||
| Tissue factor pathway inhibitor 1-alpha (TFPI) affinity | Endothelial cells (umbilical vein), human | Flow cytometry assay | IC-50 |
| Tissue factor pathway inhibitor 1-alpha (TFPI) affinity | Human factor | ELISA assay | IC-50 |
| Tissue factor pathway inhibitor 1-alpha (TFPI) affinity | Human protein | ELISA assay | IC-50 |
| Tissue factor pathway inhibitor 1-alpha (TFPI) affinity | Human protein | Surface plasmon resonance assay | IC-50 |
| Tissue factor pathway inhibitor 1-alpha (TFPI) affinity | Human protein | Kd | |
| Tissue factor pathway inhibitor 1-alpha (TFPI) affinity | Mouse protein | ELISA assay | IC-50 |
| Tissue factor pathway inhibitor 1-alpha (TFPI) affinity | Mouse protein | Surface plasmon resonance assay | IC-50 |
| Tissue factor pathway inhibitor 1-alpha (TFPI) affinity | Recombinant human factor | ELISA assay | IC-50 |
| Tissue factor pathway inhibitor 1-alpha (TFPI) affinity | Radioactivity assay | IC-50 | |
| Tissue factor pathway inhibitor 1-alpha (TFPI) affinity | IC-50 | ||
| Tissue factor pathway inhibitor 1-alpha (TFPI) inhibition | Plasma, monkey | IC-50 | |
| Tissue factor pathway inhibitor 1-alpha/Coagulation factor VIIa/Coagulation factor Xa interaction, inhibition | Endothelial cells (umbilical vein), human | ELISA assay | IC-50 |
| Tissue factor pathway inhibitor 1-alpha/Coagulation factor VIIa/Coagulation factor Xa interaction, inhibition | Human factor | ELISA assay | IC-50 |
Triggering Receptor Expressed On Myeloid Cells Like 1 (TREM-Like 1) plays a key role in platelet activation and immune responses relevant to hemophilia pathology and therapy. Testing TREM-Like 1 interactions is crucial for evaluating hemophilia drug efficacy and safety. Our service uses ELISA assays to quantify TREM-Like 1 binding, providing precise measurement of the dissociation constant (Kd), a critical parameter for assessing drug-target affinity and optimizing hemophilia treatments.
| Pharmacological Activity | Material | Method | Parameter |
|---|---|---|---|
| Trem-like transcript 1 protein (TLT-1) affinity | Recombinant human receptor | ELISA assay | Kd |
Make Order
Experimental Scheme
Implementation
Conclusion