IVD Human

Isovaleryl Coenzyme A Dehydrogenase Human Recombinant
Shipped with Ice Packs
In Stock

Description

IVD Gene: Isovaleryl-CoA Dehydrogenase

The IVD gene (OMIM: 607036) encodes the mitochondrial enzyme isovaleryl-CoA dehydrogenase, which catalyzes the third step in leucine catabolism. Its dysfunction leads to isovaleric acidemia, a rare genetic disorder characterized by neurotoxic metabolite accumulation .

Key Features of the IVD Gene

AttributeDetails
FunctionConverts isovaleryl-CoA to 3-methylcrotonyl-CoA in leucine degradation .
Chromosomal Location15q14-q15
DisordersIsovaleric acidemia (OMIM: 243500)
Pathogenic Mutations>100 identified mutations, including missense, nonsense, and splicing variants .

Isovaleric Acidemia

This autosomal recessive disorder manifests with:

  • Sweaty foot odor due to isovaleric acid accumulation.

  • Neurological symptoms: Lethargy, seizures, coma.

  • Metabolic crises triggered by infections or fasting .

In Vitro Diagnostics (IVDs)

IVDs are medical devices used to analyze human biological samples (e.g., blood, tissue) for diagnostic, monitoring, or therapeutic purposes. Their applications span infectious diseases, oncology, and genetic testing .

Applications of IVDs in Human Health

FieldExamplesTechnologies
Infectious DiseasesCOVID-19, HIV, tuberculosisPCR, ELISA, NGS
OncologyCancer biomarker detection (e.g., HER2, BRCA1)IHC, FISH, qPCR
ToxicologyDrug abuse screening (opioids, cannabinoids)Immunoassays, LC-MS/MS
Genetic TestingPrenatal screening (Down syndrome), pharmacogenomics (CYP450 variants)Microarray, Sanger sequencing

Regulatory Frameworks

IVDs are governed by stringent regulations to ensure safety and efficacy:

  • EU: IVDR (In Vitro Diagnostic Regulation) mandates clinical performance studies and Eudamed reporting .

  • USA: FDA classifies IVDs as Class I, II, or III devices, requiring premarket clearance/approval .

  • Clinical Studies: Investigational IVDs require IRB oversight and may necessitate IDE (Investigational Device Exemption) submissions .

Spatial Transcriptomics in Intervertebral Discs (IVD)

Recent single-cell RNA sequencing (scRNA-seq) of human intervertebral discs identified distinct cell populations:

  • Notochordal progenitor cells (NPPCs): Express PDGFRA and PROCR, critical for disc regeneration .

  • Chondrocytes: Subpopulations (Chond1, Chond2) show divergent ECM synthesis and signaling roles .

Cell TypeMarkersFunction
NPPCsPDGFRA, PROCRRegulate disc homeostasis
Chond1COL2A1, SOX9ECM remodeling
Chond2COL6A1, COL11A2Matrix organization

Clinical Implications

These findings highlight therapeutic targets for degenerative disc disease (DDD) .

Challenges and Future Directions

  • Precision Medicine: IVDs enable tailored treatments but require robust clinical utility data for reimbursement .

  • Global Harmonization: Disparities in IVD regulations (e.g., EU IVDR vs. FDA) complicate international trials .

  • Ethical Considerations: Use of IVDs in companion diagnostics raises concerns about informed consent and risk stratification .

Product Specs

Introduction
Isovaleryl-CoA dehydrogenase (IVD) is a mitochondrial enzyme involved in the breakdown of the amino acid leucine. Specifically, it catalyzes the third step in this pathway, converting isovaleryl-CoA to 3-methylcrotonyl-CoA. Deficiency in IVD prevents the normal breakdown of leucine, leading to a buildup of isovaleric acid in the body. This condition, known as isovaleric acidemia, can cause significant health problems due to the toxic effects of isovaleric acid on the nervous system. IVD is a homotetrameric enzyme, meaning it is composed of four identical subunits. Each subunit contains a flavin adenine dinucleotide (FAD) molecule, which is essential for its enzymatic activity.
Description
This product consists of the human IVD enzyme, produced recombinantly in E. coli bacteria. It is engineered without any glycosylation (addition of sugar molecules) and exists as a single polypeptide chain of 415 amino acids. This includes amino acids 33 to 426 of the native human IVD protein, along with an additional 20 amino acid histidine tag at the N-terminus to facilitate purification. The purified protein has a molecular weight of approximately 45.3 kDa.
Physical Appearance
Clear and colorless liquid, sterilized by filtration.
Formulation
The IVD protein is supplied in a solution at a concentration of 1 mg/ml. The solution is buffered with 20mM Tris-HCl at pH 8 and contains 1mM DTT to prevent protein degradation, and 10% glycerol to enhance stability.
Stability
For short-term storage (up to 4 weeks), keep the vial refrigerated at 4°C. For longer storage, freeze the product at -20°C. Adding a carrier protein like albumin (HSA or BSA) to a final concentration of 0.1% is recommended for long-term storage. Avoid repeated freezing and thawing of the protein solution.
Purity
The purity of the IVD protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
FLJ12715, isovaleryl-CoA dehydrogenase mitochondrial, FLJ34849, EC 1.3.99.10, IVD, ACAD2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MHSLLPVDDA INGLSEEQRQ LRQTMAKFLQ EHLAPKAQEI DRSNEFKNLR EFWKQLGNLG VLGITAPVQY GGSGLGYLEH VLVMEEISRA SGAVGLSYGA HSNLCINQLV RNGNEAQKEK YLPKLISGEY IGALAMSEPN AGSDVVSMKL KAEKKGNHYI LNGNKFWITN GPDADVLIVY AKTDLAAVPA SRGITAFIVE KGMPGFSTSK KLDKLGMRGS NTCELIFEDC KIPAANILGH ENKGVYVLMS GLDLERLVLA GGPLGLMQAV LDHTIPYLHV REAFGQKIGH FQLMQGKMAD MYTRLMACRQ YVYNVAKACD EGHCTAKDCA GVILYSAECA TQVALDGIQC FGGNGYINDF PMGRFLRDAK LYEIGAGTSE VRRLVIGRAF NADFH.

Q&A

What constitutes a "human subject" in IVD research?

Under FDA's medical device regulations, a subject in IVD research is defined as "a human who participates in an investigation, either as an individual on whom or on whose specimen an investigational device is used or as a control." This definition has important implications for researchers working with biospecimens. Even if you have no direct interaction with the individuals from whom samples were collected and possess no identifiable information about the donors, your study still qualifies as human subjects research if it uses human biospecimens with an investigational device . This regulatory classification applies regardless of whether the samples are fully de-identified, as the FDA's definition focuses on the use of the investigational device on human-derived materials rather than on identifiability of the source .

For research planning purposes, this means that protocols using any human specimens with investigational IVDs must account for human subject protections, including appropriate IRB review and, depending on risk level, potentially informed consent requirements. Researchers should not assume that using "leftover" or banked specimens exempts their work from these considerations . The methodological approach should include early consultation with your institution's IRB to determine the specific requirements applicable to your research design.

When does IVD research require IRB review?

Methodologically, researchers should approach IRB requirements by:

  • Determining whether the IVD is investigational or already approved/cleared for the intended use

  • Assessing risk level in consultation with the IRB

  • Identifying whether the study qualifies for IDE exemption

  • Preparing documentation that addresses human subject protections even when using banked specimens

Studies using investigational IVDs that present significant risk or that do not qualify for IDE exemption will require full IRB review and IDE submission to the FDA . The key methodological consideration is to engage with regulatory experts early in the research planning process to identify the appropriate regulatory pathway.

What is the difference between analytical validity and clinical validity in IVD research?

Analytical validity and clinical validity represent two distinct but essential aspects of IVD evaluation, each requiring specific methodological approaches:

Analytical Validity: This refers to how well the test can measure or detect the target analyte or biomarker of interest. Analytical validity focuses on the technical performance characteristics of the test itself . Methodologically, establishing analytical validity involves:

  • Precision evaluation (repeatability and reproducibility)

  • Cross-reactivity assessment

  • Measuring interval determination

  • Accuracy (trueness) evaluation against a comparator device

  • Determination of analytical sensitivity and specificity for detecting specific biomarkers/analytes

Clinical Validity: This assesses the ability of the test to accurately classify a patient into a disease or prognosis category. Clinical validity evaluates how well the test's results correspond to the clinical condition it aims to identify . The methodological approach includes:

  • Estimation of clinical performance measures

  • For binary qualitative tests: determining clinical sensitivity, specificity, positive and negative likelihood ratios, and predictive values

  • For qualitative tests with multiple outputs: establishing pre-test risk, post-test risks, likelihood ratios, and percentage of patients for each output

The primary distinction is that analytical validity addresses the technical accuracy of the test, while clinical validity addresses its medical accuracy in identifying the clinical condition of interest. Both are necessary components of a complete IVD evaluation framework, and researchers should design separate protocols to establish each type of validity.

What types of studies are needed to demonstrate analytical validity for IVDs?

Demonstrating analytical validity for IVDs requires a systematic series of studies addressing different performance aspects. Based on established frameworks, the following studies are typically required:

  • Precision Studies: These evaluate the test's ability to provide consistent results when repeated under varying conditions.

    • Repeatability: Assesses precision under the same operating conditions over a short time interval, including the same operators, same operating conditions, and same location

    • Reproducibility: Evaluates precision under different conditions, including different locations, operators, and instruments

  • Accuracy Studies: These compare the test results to a reference standard or comparator device to determine how close the measured values are to the true values.

  • Analytical Sensitivity Studies: These determine the lowest concentration of analyte that can be reliably detected.

  • Analytical Specificity Studies: These evaluate potential interference from structurally or physiologically related substances and assess cross-reactivity.

  • Measuring Interval Evaluation: This establishes the range of analyte values that can be directly measured without dilution or concentration.

The methodological approach should follow established guidelines such as those in CLSI documents EP05-A3 and EP12-A2, which provide detailed frameworks for study design and analysis . Additionally, FDA has issued specific guidance documents for particular types of IVDs (e.g., HIV tests, companion diagnostics, HPV tests) that should be consulted when designing analytical validation studies for these specific applications .

How are IVDs classified based on their intended use?

IVDs are classified based on their intended use, which determines both the regulatory pathway and the type of evidence required for validation. The primary classifications include:

  • Diagnostic IVDs: Tests designed to confirm the presence of a specific disease or condition. These require demonstration of high clinical sensitivity and specificity relative to a clinical reference standard .

  • Aid in Diagnosis IVDs: Tests that provide information to be considered along with other clinical findings. These typically have less stringent performance requirements than standalone diagnostic tests .

  • Screening IVDs: Tests used to detect a disease or condition in asymptomatic individuals. These prioritize high sensitivity to minimize false negatives, with confirmation by more specific tests .

  • Monitoring IVDs: Tests used to track disease progression or treatment effects over time. These emphasize precision and reproducibility to detect meaningful changes .

  • Predisposition (Risk Assessment) IVDs: Tests that evaluate the likelihood of developing a disease or condition. These require long-term studies correlating test results with clinical outcomes .

  • Prognostic IVDs: Tests that predict the likely course of a disease. These require evidence linking test results to clinical outcomes without intervention .

  • Treatment Response (Prediction) IVDs: Tests that predict response to specific treatments, including companion diagnostics. These require evidence of the test's ability to identify patients likely to benefit from specific therapeutic interventions .

Each classification has specific design considerations for clinical validation based on the intended use, which affects study endpoints, comparator selection, and performance targets. Methodologically, researchers should clearly define the intended use category early in development, as this determines the appropriate validation strategy.

How can researchers develop effective in vitro models for testing IVD efficacy?

Developing effective in vitro models for testing IVD efficacy requires careful consideration of physiological relevance, particularly when the in vivo environment significantly influences biomarker expression or detection. Based on recent methodological advances, researchers should consider the following approach:

For models involving tissue-based diagnostics (such as intervertebral disc tissue), maintaining physiologically relevant conditions is critical. Researchers have successfully developed in vitro culture systems that maintain metabolically active tissue within a loading environment similar to that experienced in the human body . Such models have proven valuable for testing the efficacy of cell-based and tissue-engineering therapies.

The methodological workflow should include:

  • Physiological Environment Replication: Design culture systems that mimic relevant in vivo conditions. For example, when testing IVDs for musculoskeletal applications, incorporate appropriate mechanical loading regimes based on measurements taken during typical human activities .

  • Tissue Integrity Verification: Implement regular assessments of tissue viability and integrity throughout the culture period. Successful models have demonstrated maintenance of metabolic cell activity, proteoglycan content, and cellular phenotype for up to two weeks .

  • Comparative Controls: Always include unloaded controls to determine the specific effects of the physiological environment. Research has shown that loaded tissue cultures often demonstrate improved parameter maintenance compared to unloaded controls, highlighting the importance of mimicking in vivo conditions .

  • Validation Metrics: Establish clear metrics for validating model effectiveness, including:

    • Metabolic activity measurements

    • Cellular phenotype stability assessment

    • Tissue integrity evaluation

    • Proteoglycan content preservation

This methodological approach creates an invaluable platform for investigating IVD efficacy under conditions that more accurately reflect the human physiological environment, potentially improving the translation of in vitro findings to clinical applications.

What methodologies are recommended for handling data contradictions in IVD research?

Data contradictions represent a significant challenge in IVD research, particularly when dealing with complex, interdependent data items. Effective methodological approaches to handling such contradictions involve structured representation and evaluation:

Contradiction Pattern Notation: A formalized approach to representing contradiction patterns uses three key parameters (α, β, θ) :

  • α: Number of interdependent items

  • β: Number of contradictory dependencies defined by domain experts

  • θ: Minimal number of required Boolean rules to assess these contradictions

This notation helps bridge the gap between biomedical domain knowledge (required for defining contradictions) and informatics domain knowledge (needed for efficient implementation in assessment tools) .

Methodological Approach to Handling Contradictions:

  • Structured Classification: Develop a structured classification of contradiction checks that captures the complexity of the interdependencies. This enables more effective implementation of contradiction assessment frameworks across multiple domains .

  • Boolean Minimization: Apply Boolean minimization techniques to reduce the number of rules required for contradiction assessment. Research in biobank and COVID-19 domains has shown that the minimum number of Boolean rules (θ) can be significantly lower than the number of contradictions (β) described by domain experts .

  • Domain-Specific Rule Development: Collaborate with domain experts to identify and formalize contradiction rules specific to the IVD application area. This ensures that data quality assessments are both computationally efficient and scientifically meaningful .

  • Implementation in Assessment Frameworks: Incorporate the formalized rules into data quality assessment frameworks, using the (α, β, θ) notation to classify and prioritize contradiction checks .

This structured approach to handling contradictions helps manage the complexity of multidimensional interdependencies within health datasets, improving data quality for IVD research applications.

What design considerations should be made for clinical validation based on different IVD intended uses?

Clinical validation design must be tailored to the specific intended use of the IVD to ensure appropriate evidence generation. The methodological approach varies significantly across intended use categories:

  • Diagnostic IVDs:

    • Study Design: Case-control or cohort studies comparing test results to a clinical reference standard

    • Key Metrics: Clinical sensitivity, specificity, positive predictive value, negative predictive value

    • Special Considerations: Spectrum of disease severity must be adequately represented; prevalence in the study population should reflect the intended use population

  • Aid in Diagnosis IVDs:

    • Study Design: Typically evaluate the added value of the test when used alongside other clinical information

    • Key Metrics: Incremental value measurements, changes in diagnostic confidence

    • Special Considerations: Should demonstrate additional diagnostic value beyond existing methods

  • Screening IVDs:

    • Study Design: Large cohort studies in asymptomatic populations

    • Key Metrics: Prioritize sensitivity, negative predictive value, and positive predictive value in low-prevalence settings

    • Special Considerations: False positive management strategy is critical; cost-effectiveness analyses often required

  • Monitoring IVDs:

    • Study Design: Longitudinal studies with repeated measurements

    • Key Metrics: Precision, reproducibility, minimal detectable change

    • Special Considerations: Must establish clinically significant thresholds for changes in measurements

  • Predisposition/Risk Assessment IVDs:

    • Study Design: Long-term prospective cohort studies or case-control studies with follow-up

    • Key Metrics: Relative risk, odds ratio, area under ROC curve

    • Special Considerations: Time-to-event analyses often relevant; need to account for competing risks

  • Prognostic IVDs:

    • Study Design: Prospective cohort studies with defined clinical endpoints

    • Key Metrics: Survival analysis metrics, hazard ratios

    • Special Considerations: Must control for standard prognostic factors to demonstrate independent value

  • Treatment Response (Prediction) IVDs:

    • Study Design: Randomized controlled trials or well-designed retrospective analyses

    • Key Metrics: Treatment effect modification, interaction tests

    • Special Considerations: Should demonstrate differential treatment effect based on test results

For multi-analyte assays with algorithmic analyses (MAAA) or in vitro diagnostics multivariate index assays (IVDMIA), additional validation of the algorithm itself is required, including verification of computational reproducibility and assessment of overfitting risks .

The methodological approach should include pre-specification of all clinical validation study elements, including endpoints, analysis plans, and success criteria, tailored to the specific intended use category.

How can researchers develop evidence of clinical utility that satisfies regulatory requirements and payer considerations?

Developing evidence of clinical utility for IVDs requires a strategic approach that addresses both regulatory requirements and payer considerations. Clinical utility refers to the demonstration that a test improves health outcomes when used as intended and provides value to patients, clinicians, and healthcare systems .

Methodological Framework for Clinical Utility Evidence Development:

  • Demonstrate Positive Patient Outcomes:

    • Primary approach: Design studies that directly measure the impact of test-directed interventions on patient outcomes

    • Alternative approach: Develop a chain of evidence linking test results to established interventions with known benefits

  • Link Tests to Clinical Care Decisions:

    • Document how test results inform specific clinical decisions

    • Demonstrate that these decisions lead to improved patient management

    • Quantify the clinical impact of testing-informed decisions versus standard care

  • Strategic Study Design Selection:

    • Randomized controlled trials: Gold standard for demonstrating direct impact on outcomes

    • Decision analysis models: Can incorporate existing clinical data to simulate long-term outcomes

    • Observational studies: May be appropriate when randomization is not feasible

    • Health economic evaluations: Address cost-effectiveness and resource utilization questions

  • Stratified Clinical Utility Assessment by Test Type:

    • Diagnostic tests: Focus on time to diagnosis, avoided procedures, improved treatment selection

    • Screening tests: Emphasize early detection benefits, mortality reduction, quality of life impacts

    • Monitoring tests: Demonstrate improved disease management, reduced complications

    • Predictive/Prognostic tests: Show improved risk stratification leading to optimized treatment decisions

  • Address Payer-Specific Evidence Requirements:

    • Beyond regulatory approval, develop evidence specifically addressing payer decision frameworks

    • Include comparative effectiveness against existing methods

    • Incorporate health economic analyses appropriate to the healthcare system context

    • Consider real-world evidence to supplement clinical trial data

By employing this comprehensive framework, researchers can develop evidence that supports both regulatory approval and coverage decisions, addressing the full spectrum of stakeholder requirements for clinical utility demonstration.

What are the specific considerations for multi-analyte assays with algorithmic analyses in IVD research?

Multi-Analyte Assays with Algorithmic Analyses (MAAA) or In Vitro Diagnostics Multivariate Index Assays (IVDMIA) present unique methodological challenges in IVD research due to their complexity. These tests analyze multiple variables using an algorithm to generate a single, patient-specific result . Researchers should consider the following methodological approaches:

The methodological rigor required for MAAA/IVDMIA validation exceeds that of single-analyte tests, particularly regarding computational reproducibility, algorithm transparency, and the need to demonstrate that the multivariate approach provides value beyond simpler testing strategies.

Product Science Overview

Structure and Function

Isovaleryl Coenzyme A Dehydrogenase is encoded by the IVD gene located on chromosome 15 in humans . The enzyme is a member of the acyl-CoA dehydrogenase family, which is characterized by its ability to catalyze the dehydrogenation of acyl-CoA derivatives. The enzyme’s active site binds flavin adenine dinucleotide (FAD) as a cofactor, which is essential for its catalytic activity .

The primary function of IVD is to facilitate the breakdown of leucine, an essential amino acid, by catalyzing the conversion of isovaleryl-CoA to 3-methylcrotonyl-CoA. This reaction is a critical step in the leucine catabolic pathway, which ultimately leads to the production of acetyl-CoA and acetoacetate, molecules that can be utilized for energy production .

Genetic Deficiency and Associated Disorders

A deficiency in IVD activity due to mutations in the IVD gene results in a metabolic disorder known as isovaleric acidemia . This condition is characterized by the accumulation of isovaleric acid, a toxic compound that can lead to severe metabolic disturbances. Symptoms of isovaleric acidemia can range from mild to life-threatening and may include vomiting, lethargy, seizures, and a distinctive “sweaty feet” odor .

Isovaleric acidemia can present in two major clinical forms: an acute neonatal form and a chronic intermittent form. The acute form typically manifests shortly after birth with severe metabolic crises, while the chronic form presents later in life with intermittent episodes of metabolic decompensation .

Recombinant Production

The recombinant form of Isovaleryl Coenzyme A Dehydrogenase is produced using genetic engineering techniques. The IVD gene is cloned into an expression vector, which is then introduced into a suitable host organism, such as Escherichia coli or yeast. The host cells are cultured under conditions that promote the expression of the recombinant enzyme, which is subsequently purified using chromatographic techniques .

Recombinant IVD is used in various research applications, including studies on enzyme kinetics, structure-function relationships, and the development of therapeutic strategies for isovaleric acidemia. The availability of human recombinant IVD allows for detailed biochemical and biophysical analyses, which are essential for understanding the enzyme’s function and its role in metabolic disorders .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.