Discovery & Development Services for Cardiovascular Diseases
Discovery & Development
Online Inquiry
* Please note that all of our services and products are intended for preclinical research use only and cannot be used to diagnose, treat or manage patients.

Discovery & Development Services for Cardiovascular Diseases

Inquiry

One-Stop Shop for Cardiovascular Drug Development and Preclinical Studies

Ace Therapeutics is a contract research organization engaged in cardiovascular disease research. We aim to provide comprehensive solutions and R&D outsourcing services for our clients worldwide to accelerate cardiovascular drug discovery and development.

Get Free Quote
Cardiovascular-Drug-Development-and-Preclinical-Studies

Challenges in Cardiovascular Drug Development

The development of novel therapeutics for cardiovascular disease confronts multiple translational challenges.

  • The inherent complexity of cardiovascular pathophysiology with its diverse disease subtypes and multifactorial mechanisms.
  • The limited clinical predictivity of current preclinical models that often fail to recapitulate human disease progression.
  • The persistently high attrition rates in clinical development stemming from insufficient therapeutic efficacy or unacceptable safety profiles.
Challenges-in-Cardiovascular-Drug-Development

Animal Model Development Services

We understand the difficulties of the current research into cardiovascular disease, and are committed to providing animal model construction services, conventional genetically engineered animal models, and customized functional animal diets for the basic research and drug development of cardiovascular diseases.

img-arrow

Animal Fetching

  • Rat
  • Pigs
  • Mice
  • Dogs
  • Rabbits
  • Non-human Primates

Model Construction

  • Environmental Stress
  • Surgery
  • Foods/Drugs

Model Evaluation

  • Blood Serum Analysis
  • Anatomical Validation
  • Histological Validation
Animal-Model-Development

Given the complex pathophysiology and chronic progression of atherosclerosis, well-characterized animal models that faithfully recapitulate key aspects of human disease pathogenesis have become indispensable tools for investigating disease mechanisms and evaluating novel therapeutic interventions. Recent advances in preclinical model development have yielded increasingly sophisticated systems that more accurately mimic human atherosclerotic progression, enabling rigorous investigation of disease pathogenesis and accelerating the discovery of innovative treatment strategies.

  • Genetically Manipulated Models of Atherosclerosis
  • High-Fat Diet Induced Models of Atherosclerosis
  • LPS-induced Animal Models of Atherosclerosis
  • Surgery-induced Animal Models of Atherosclerosis

Well-established animal models of hypertension serve as essential tools for investigating hypertensive disorders and their associated complications, with two primary classifications. Genetic hypertension models, which develop elevated blood pressure without external manipulation, provide valuable systems for studying the fundamental mechanisms of essential hypertension and evaluating novel antihypertensive therapies. Experimentally induced hypertension models, created through pharmacological, dietary, or surgical interventions, offer rapid induction timelines and are particularly suited for investigating secondary hypertension mechanisms triggered by specific environmental or pharmacological factors.

  • Essential Hypertension Models
  • Drug Induced Hypertension Models
  • Environmental Induced Hypertension Models
  • Surgical Induced Hypertension Models
  • Genetically Engineered Hypertension Models

Thrombosis, characterized by pathological intravascular clot formation, represents a critical mechanism underlying major cardiovascular and cerebrovascular events, including acute myocardial infarction and ischemic stroke, through its dual manifestations as arterial and venous thromboembolism. Given the central role of thrombotic processes in numerous life-threatening conditions, well-validated animal models of thrombosis have become indispensable for elucidating disease mechanisms and advancing the development of novel antithrombotic therapies with improved efficacy and safety profiles.

  • Inferior Vena Cava Ligation Induced Thrombosis Models
  • Inferior Vena Cava Stenosis Induced Thrombosis Models
  • Mechanical Injury Induced Thrombosis Models
  • Pro-coagulant Intervention Induced Thrombosis Models
  • Ferric Chloride (FeCl3) Induced Thrombosis Models

The myocardial ischemia-reperfusion arrhythmia model serves as a well-established preclinical system for investigating ischemic cardiomyopathy pathophysiology, wherein transient coronary occlusion followed by controlled reperfusion induces characteristic electrophysiological disturbances that mimic clinical manifestations of ischemic heart disease. This model specifically recapitulates the pathophysiological cascade of myocardial injury resulting from temporary blood flow interruption and subsequent restoration, providing critical insights into ischemia-reperfusion injury mechanisms and potential therapeutic interventions.

  • Coronary Artery Ligation Induced Myocardial Ischemia-Reperfusion Injury (I/R) Models

Many existing antiarrhythmic agents paradoxically exhibit proarrhythmic potential or induce other cardiovascular complications, highlighting the critical need for developing safer and more effective therapeutic alternatives. The establishment of physiologically relevant arrhythmia models is fundamental to this endeavor, with murine models (mice and rats) serving as the predominant preclinical systems due to their conserved cardiac anatomy and electrophysiological properties that closely mirror human pathophysiology, making them indispensable for investigating arrhythmia mechanisms and evaluating novel pharmacological interventions.

  • Drug Induced Arrhythmias Models
  • Electrical Stimulation Induced Arrhythmias Models
  • Surgery Induced Arrhythmias Models

Heart failure arises from diverse etiologies that ultimately impair myocardial contractility, leading to insufficient cardiac output to meet systemic metabolic demands and subsequent development of characteristic clinical manifestations. To recapitulate this complex pathophysiology, researchers have established multiple preclinical models through distinct induction methods, including pharmacological cardiotoxicity, chronic metabolic stress from high-fat diets, and surgical hemodynamic overload via vascular constriction, each exhibiting unique patterns of cardiac functional deterioration and remodeling that mirror specific clinical heart failure phenotypes.

  • Essential Heart Failure Animal Models
  • High-Fat Diet Induced Heart Failure Models
  • Drug Induced Heart Failure Models
  • Surgery Induced Heart Failure Models

Hypertrophic cardiomyopathy represents a maladaptive myocardial response to sustained pathological stimuli, characterized by cardiomyocyte hypertrophy and architectural remodeling. To investigate this complex pathophysiology, researchers have established multiple validated preclinical models that accurately recapitulate disease features, including pressure-overload models induced by surgical aortic constriction, pharmacological models utilizing cardiotropic agents, and genetically engineered models with targeted mutations.

  • Pressure Overloading Method Induced Hypertrophic Cardiomyopathy Models
  • Volumetric Loading Method Induced Hypertrophic Cardiomyopathy Models
  • Myocardial Infarction Method Induced Hypertrophic Cardiomyopathy Models
  • Chemicals/Drugs Induced Hypertrophic Cardiomyopathy Models

Heart valve disease pathogenesis involves intricate inflammatory and apoptotic pathways, though the precise molecular mechanisms remain incompletely characterized, necessitating the development of physiologically relevant animal models to elucidate disease progression. An optimal preclinical model of valvular pathology should combine rapid induction kinetics with hallmark features of valve inflammation and fibrosis, while demonstrating quantifiable structural abnormalities through advanced imaging modalities to facilitate comprehensive mechanistic and therapeutic investigations.

  • Bacterial-Induced Heart Valve Disease Models
  • Diet-Induced Heart Valve Disease Models
  • Drug-Induced Heart Valve Disease Models

Cardiovascular infections caused by diverse pathogens including bacteria, viruses, mycoplasma, chlamydia, and rickettsia can manifest as distinct clinical entities such as infective endocarditis, myocarditis, pericarditis, valvulopathies, and systemic vasculitis. Contemporary research utilizes pathogen-specific animal models, with enterovirus-infected models (particularly coxsackievirus B3 and EV71 strains) effectively mimicking the clinical progression of viral myocarditis, enabling mechanistic studies of host-pathogen interactions and therapeutic development.

  • Animal Models of Infective Endocarditis
  • Animal Models of Infectious Myocarditis
  • Animal Models of Systemic Infection

Preclinical Cardiovascular Drug Development Services

Ace Therapeutics specializes in the preclinical development of novel cardiovascular therapeutics tailored to disease mechanisms, drug modalities, and specific therapeutic targets. Our end-to-end solutions ensure optimized efficacy, safety, and translational potential for your candidate.

Drug Discovery by Therapeutic Areas
Durg Discovery by Modality
Durg Discovery by Targets

Preclinical Research Services for Cardiovascular Drugs

At Ace Therapeutics, we deliver end-to-end preclinical research services for cardiovascular drug development, encompassing rigorous in vitro and in vivo evaluations across pharmacodynamics, safety pharmacology, toxicology, and pharmacokinetics to generate robust datasets that inform clinical trial design and risk assessment.

In-Vitro-Pharmacodynamic-Evaluation

In Vitro Pharmacodynamic Evaluation of Cardiovascular Drugs

Our comprehensive in vitro pharmacodynamic evaluation platform enables systematic characterization of drug mechanisms and pharmacological effects during early discovery phases, providing critical data to de-risk cardiovascular drug development through predictive assessment of therapeutic potential and target engagement.

  • Assessment of Target/Receptor Binding Affinity
  • Assessment of Antibody Binding Affinity
  • Functional Assessment of Cell Cardiomyocytes
  • Functional Assessment of Isolated Hearts
  • hERG Safety Assessment
In-Vivo-Pharmacodynamic-Evaluation

In Vivo Pharmacodynamic Evaluation of Cardiovascular Drugs

Our specialized in vivo pharmacodynamic evaluation platform enables direct measurement of critical drug activity parameters in physiologically relevant systems, providing essential preclinical data to characterize therapeutic efficacy, dose-response relationships, and mechanism of action for cardiovascular drug candidates.

In-Vitro-Pharmacokinetics-Evaluation

In Vitro Pharmacokinetics Evaluation of Cardiovascular Drugs

Achieving an ideal pharmacokinetic profile represents a critical determinant of clinical success for drug candidates. Our advanced in vitro pharmacokinetic technology platform delivers scientifically validated solutions for characterizing key ADME parameters of cardiovascular therapeutics, enabling data-driven optimization of drug candidates' pharmacokinetic properties to enhance therapeutic potential and clinical translatability.

  • In Vitro Physicochemical Property Assessment
  • Cell Permeability & Transporter Assessment
  • In Vitro Drug Metabolic Stability Assessment
  • Metabolism-Mediated Drug-Drug Interaction Assessment
In-Vivo-Pharmacokinetics-Evaluation

In Vitro Pharmacodynamic Evaluation of Cardiovascular Drugs

Prior to initial human administration, comprehensive assessment of a drug candidate's pharmacokinetic profile through validated animal studies is essential to ensure safe and effective clinical translation. Our established preclinical platform provides robust in vivo pharmacokinetic evaluation services specifically optimized for cardiovascular therapeutics, delivering critical data on absorption, distribution, metabolism and excretion (ADME) parameters to support regulatory submissions and clinical trial design.

General-Toxicity-Evaluation

General Toxicity Evaluation of Cardiovascular Drugs

Systematic general toxicity evaluation constitutes a critical component of the preclinical safety assessment for cardiovascular therapeutics, encompassing comprehensive characterization of acute, subchronic, and chronic toxicity profiles. Our preclinical platform delivers end-to-end general toxicity evaluation services specifically designed for cardiovascular drugs.

  • Acute Toxicity Testing (14-day Observation)
  • Subchronic Toxicity Evaluation (90 Day Studies)
  • Chronic Toxicity Evaluation (6–12 Month Studies)
General-Toxicity-Evaluation

Genetic Toxicity Evaluation of Cardiovascular Drugs

Genotoxicity represents the deleterious effects on genetic material including DNA damage and chromosomal aberrations, a particular concern for cardiovascular therapeutics due to their typically chronic administration regimens. Our preclinical genotoxicity evaluation platform provides comprehensive assessment of potential mutagenic risks for cardiovascular drug candidates through validated in vitro and in vivo studies, enabling early identification of genetic safety concerns during lead optimization and preclinical development phases.

  • Ames Test
  • In Vitro Mammalian Chromosome Aberration Test
  • Micronucleus Assay
Reproductive-Toxicity-Evaluation

Reproductive Toxicity Evaluation of Cardiovascular Drugs

Reproductive toxicity evaluation provides essential safety data required for clinical trial authorization and regulatory approval, establishing critical risk-benefit profiles for drugs with potential effects on fertility, embryonic development, and postnatal growth. Our reproductive toxicity testing platform delivers complete evaluation services for cardiovascular therapeutics.

  • Fertility and Early Embryonic Development (FEED) Tests
  • Embryo-Fetal Development (EFD) Tests
  • Perinatal Development (PPND) Tests
Safety-Pharmacology-Evaluation

Safety Pharmacology Evaluation of Cardiovascular Drugs

Safety pharmacology evaluation provides critical insights into potential adverse physiological effects of novel cardiovascular drugs at therapeutic and supratherapeutic doses, enabling early identification of functional risks to vital organ systems. Our safety pharmacology platform delivers comprehensive cardiovascular assessments, including:

  • Core Battery Safety Pharmacology Studies (Cardiovascular System, Respiratory System, and Central Nervous System Evaluations)
  • Supplemental Safety Pharmacology Studies (Urinary System, Autonomic Nervous System, Gastrointestinal System Evaluations)

Why Partner with Us?

Ace Therapeutics provides scientifically validated, regulatory-ready solutions that bridge discovery research and clinical development, delivering robust preclinical data packages to accelerate your cardiovascular therapeutics while maintaining the highest standards of scientific rigor and regulatory compliance.

OurAdvantages
  • Advanced Biotechnology
  • Customizable Design
  • Competitive Pricing
  • Best After-sales Service

FAQ for Cardiovascular Drug Development

Q: What’s your approach to repurposing drugs for CVD?

A: We leverage computational biology and network pharmacology to identify repurposing candidates, followed by rapid in vitro / in vivo validation in relevant CVD models.

Q: Do you support gene therapy or RNA-based CVD therapeutics?

A: Yes, from AAV vector design (e.g., for familial hypercholesterolemia) to LNP-formulated siRNA/mRNA delivery (e.g., targeting PCSK9).

Q: How do you address the translatability gap between preclinical models and human CVD?

A: We employ a tiered model strategy, combining genetically engineered rodents, large-animal models (e.g., porcine MI models), and human iPSC-derived cardiomyocytes/3D tissue constructs to improve clinical relevance.

Q: How can small molecules overcome safety challenges in CVD (e.g., QT prolongation)?

A: Our early-stage screening includes multi-ion channel profiling (hERG, Nav/Cav) and structural optimization to mitigate proarrhythmic risks while maintaining efficacy.