FH Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Factor H (fH)-Targeting Monoclonal Antibodies

Factor H is a regulatory protein in the complement system, and its allelic variants (e.g., Y402H) are linked to age-related macular degeneration (AMD). Monoclonal antibodies like MBI-7 distinguish between fH-Y402 and fH-H402 variants, enabling risk stratification for AMD .

  • Diagnostic Use:

    • ELISA with MBI-7 quantifies fH variants in plasma, identifying individuals at higher AMD risk. Total fH levels average 233 mg/L in young adults and 269 mg/L in elderly populations, with elevated levels in AMD cohorts (288 mg/L) .

    • Heterozygous subgroups show balanced elevation of both fH-Y402 and fH-H402 variants, suggesting a compensatory mechanism .

Anti-fHbp Monoclonal Antibodies

fHbp, a virulence factor in Neisseria meningitidis, enables immune evasion by binding human fH. Monoclonal antibodies targeting fHbp enhance bactericidal activity via complement-mediated killing .

  • Key Antibodies:

    • JAR 41: A broadly cross-reactive murine IgG1 mAb binding fHbp variants across all three groups (1, 2, 3). Demonstrates EC50 values of 1.72 × 10⁻¹⁰ M to 0.9 × 10⁻¹¹ M .

    • Cooperative mAb pairs (e.g., non-overlapping epitopes) amplify serum bactericidal activity (SBA) by enabling hexameric IgG-C1q clustering .

AntibodyTargetCross-ReactivityApplication
JAR 41fHbp variantsGroups 1, 2, 3Meningococcal infection therapy
1A12fHbp variantsPan-variantVaccine development
  • Clinical Impact:

    • Coupled mAbs increase SBA titers by >50% in vitro, offering potential for next-generation meningococcal vaccines .

Anti-Collagen III Antibody (Clone FH-7A)

The FH-7A clone targets collagen alpha 1(III) chain (COL3A1), a structural protein implicated in Ehlers-Danlos syndrome and fibrosis .

  • Research Applications:

    • Western blot, ELISA, IHC: Detects collagen III in human/rat tissues with high specificity.

    • Therapeutic Potential: Linked to ADGRG1 signaling in neuronal migration and RhoA pathway regulation .

Therapeutic mAbs in Familial Hypercholesterolemia (FH)

While not directly targeting FH, PCSK9 inhibitors like evolocumab and alirocumab are pivotal in FH management:

  • Efficacy by Genotype:

    • HeFH: LDL-C reductions of 48–63% with residual LDLR activity .

    • HoFH: Responses depend on LDLR mutations:

      • Null/null mutations: Minimal/no response .

      • Defective mutations: LDL-C reductions up to 40.8% .

mAbTargetLDL-C Reduction (HeFH)HoFH Response
EvolocumabPCSK959–61% 13–31% (defective only)
AlirocumabPCSK948–60% 35.6% (variable)

Future Directions

  • Bispecific mAbs: Combining fHbp-targeting mAbs with immune effectors (e.g., cytokines) for enhanced efficacy .

  • Gene Silencing: siRNA therapies (e.g., inclisiran) for sustained PCSK9 inhibition in FH .

Product Specs

Buffer
PBS, pH 7.4, containing 0.02% sodium azide as a preservative and 50% Glycerol.
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
Synonyms
FH antibody; Fumarase antibody; Fumarate hydratase antibody; Fumarate hydratase mitochondrial antibody; Fumarate hydratase; mitochondrial antibody; FUMH_HUMAN antibody; HLRCC antibody; LRCC antibody; MCL antibody; MCUL 1 antibody; MCUL1 antibody; MS709 antibody; Multiple hereditary cutaneous leiomyomata antibody
Target Names
FH
Uniprot No.

Target Background

Function
FH (Fumarate Hydratase) catalyzes the reversible stereospecific interconversion of fumarate to L-malate. Research in other species has shown that specific isoforms of this protein function in defined pathways and exhibit a preference for one direction over the other (probable). FH plays a crucial role in the tricarboxylic acid (TCA) cycle, catalyzing the hydration of fumarate to L-malate. This reaction facilitates a transition step in the production of energy in the form of NADH. Additionally, FH catalyzes the dehydration of L-malate to fumarate. Fumarate metabolism in the cytosol is involved in the urea cycle and arginine metabolism, with fumarate being a by-product of these processes. FH also participates in DNA repair by promoting non-homologous end-joining (NHEJ). In response to DNA damage and phosphorylation by PRKDC, FH translocates to the nucleus and accumulates at DNA double-strand breaks (DSBs). It acts by catalyzing the formation of fumarate, an inhibitor of KDM2B histone demethylase activity. This results in enhanced dimethylation of histone H3 'Lys-36' (H3K36me2).
Gene References Into Functions
  1. Our findings suggest that over half of histologically diagnosed leiomyomata with bizarre nuclei may be associated with FH alterations or FH mutations PMID: 28863073
  2. In this study, all cases of atypical intradermal smooth muscle neoplasms and cutaneous leiomyosarcoma examined showed retained expression of FH PMID: 28703412
  3. The objective of this study was to examine common functional pathways in uterine leiomyomas (ULM) with different driver mutations. We collected ULM with MED12, HMGA2, and FH mutations and analyzed the selected markers by immunohistochemistry. PMID: 29790226
  4. Induction of ferroptosis in fumarate hydratase-inactivated tumors presents an opportunity for synthetic lethality in cancer. PMID: 29917289
  5. A case series of two sisters in their 20s who presented with isolated uterine leiomyomas and were found to carry a novel mutation for the fumarate hydratase gene. PMID: 28700432
  6. The clinical spectrum associated with hereditary leiomyomatosis and renal cell carcinoma syndrome in French FH mutation carriers has been reassessed. PMID: 28300276
  7. As with many emerging cancer syndromes, the phenotypic spectrum of hereditary leiomyomatosis and renal cell cancer (HLRCC) is likely to expand as the FH gene becomes incorporated on more multiplex germline panels. This will also help to clarify the potential involvement of other tumor types in HLRCC. PMID: 28620008
  8. A novel mutation in the FH gene (c.1349_1352delATGA) in hereditary leiomyomatosis and renal cell cancer syndrome PMID: 27566483
  9. We demonstrate that the mechanism of fumarate hydratase distribution is alternative transcription initiation from a broad promoter. PMID: 27037871
  10. Fumarate increases ferritin gene transcription by activating the NRF2 (nuclear factor [erythroid-derived 2]-like 2) transcription factor. PMID: 28289076
  11. FH-Ser75 phosphorylation level inversely correlates with the OGT level and poor prognosis in pancreatic cancer patients. This report elucidates the mechanism underlying transcription regulation by FH and the linkage between dysregulated OGT activity and growth advantage of cancer cells under glucose deficiency. PMID: 28628081
  12. Loss of FH immunohistochemical (IHC) expression in cutaneous leiomyomas is a sensitive and specific marker for detection of hereditary leiomyomatosis and renal cell carcinoma (HLRCC). FH expression by IHC was absent in 9 specimens and retained in 85 specimens. Two cases were equivocal with minimal FH expression, while succinate dehydrogenase B expression was retained in 95 specimens and equivocal in 1 specimen. PMID: 28288038
  13. LSH promoted cancer progression in part by regulating expression of fumarate hydratase (FH). PMID: 27302170
  14. Cascade FH mutation screening enabled the early diagnosis of a renal tumor in an asymptomatic parent of a child with fumarate hydratase deficiency, resulting in timely and possibly life-saving treatment. PMID: 28747166
  15. Results indicate that FH-deficient uterine leiomyomas occur with a high frequency among atypical leiomyomas and infrequently in nonatypical leiomyomas. They are often histologically distinctive and seem to have a low biological potential with no significant association with leiomyosarcoma. PMID: 27454940
  16. Mutations in some enzymes of the TCA cycle, such as Isocitrate dehydrogenase (IDH), succinate dehydrogenase (SDH), and fumarate hydratase (FH), are associated with the accumulation of metabolites that can influence various aspects of cancer development and progression. These metabolites are termed onco-metabolites. PMID: 27528759
  17. Two partial FH gene deletions were identified, with one resulting in loss of exon 1 and the upstream region of the FH gene only. Kidney cancer was diagnosed in 9 (32%) of 28 patients and 7 (54%) of 13 families possessing either complete or partial FH deletions. Cutaneous and uterine leiomyomas were observed at similar rates to those in FH point mutation families. PMID: 28196407
  18. Of the 12 available members of this second generation, 6 (1 man and 5 women, aged 44-57 years) had a novel FH mutation. All had the same mild phenotype with cutaneous asymptomatic leiomyomas, uterine fibroids (if women), and no kidney tumor. PMID: 27161211
  19. After indirect co-culture, OP was increased in the BxPc-3 and Panc-1 cells. Correspondingly, succinate dehydrogenase, FH, and MCT expression were increased. After the MCT1-specific inhibitor removed 'tumor-stromal' metabolic coupling, the migration and invasion abilities of the pancreatic cancer cells were decreased. PMID: 28260082
  20. Negative FH staining could indicate a high risk of hereditary leiomyomatosis and renal cell cancer, but it could also suggest the presence of a syndrome in up to 25% of sporadic cases. PMID: 27097334
  21. A novel deletion of the Fumarate Hydratase (FH) gene was identified in a 22-year-old male and his father, which predisposes to an aggressive form of renal cell cancer. PMID: 26275867
  22. While the great majority of patients with hereditary leiomyomatosis and renal cell carcinoma will have FH-deficient leiomyomas, 1% of all uterine leiomyomas are FH deficient, usually due to somatic inactivation. PMID: 26574848
  23. A significant association with overall survival was confirmed for the SDHC gene, SDHD gene, and FH gene. The SDHC gene and FH gene were the primary factors contributing to the different overall survival times of colorectal carcinoma. PMID: 26377099
  24. This study describes a novel mutation in the fumarate hydratase gene in a family with atypical uterine leiomyomas and hereditary leiomyomatosis and renal cell cancer. PMID: 26493120
  25. Fine mapping of the uterine leiomyoma locus on 1q43 close to a lncRNA in the RGS7-FH interval. PMID: 26113603
  26. Morphology and immunohistochemistry for fumarate hydratase aid in the detection of fumarate hydratase mutations in uterine leiomyomas from young patients. PMID: 26457356
  27. Case report of a family with a fumarate hydratase mutation causing uterine leiomyomas and renal cell cancer. PMID: 25923021
  28. These findings 1) confirm that germline FH mutations may present, albeit rarely, with pheochromocytoma or paraganglioma; and 2) extend the clinical phenotype associated with FH mutations to pediatric pheochromocytoma. PMID: 25004247
  29. A multiplex snapback primer system was developed for the simultaneous detection of JAK2 V617F and MPL W515L/K mutations in Philadelphia chromosome- (Ph-) negative myeloproliferative neoplasms (MPNs). PMID: 24729973
  30. Germline mutations in FH confer predisposition to malignant pheochromocytomas and paragangliomas. PMID: 24334767
  31. A novel missense mutation has been found in the fumarate hydratase gene in familial renal cell cancer patients lacking cutaneous leiomyomas. PMID: 24684806
  32. These results extend the range of clinical and biochemical variation associated with fumarase deficiency. PMID: 21560188
  33. Mutation in the fumarate hydratase gene is associated with hepatic diseases in twins. PMID: 24182348
  34. We report two FH gene mutations, one novel and one previously described, in two young patients with sporadic uterine fibroids and decreased fumarate hydratase activity in lymphocytes. PMID: 22764886
  35. Fumarate hydratase mRNA expression decreases significantly in correlation with the transition from normal renal parenchyma to renal cell cancer. PMID: 23295344
  36. Somatic mutations in FH do not show a pronounced effect in nonsyndromic uterine leiomyomas compared to that of their syndromic counterparts. PMID: 22528940
  37. Gastric leiomyoma was diagnosed in a 38-year-old female MCUL patient on endoscopy performed because of dyspepsia. Routine colonoscopy disclosed hyperplastic polyposis. Testing revealed a previously unreported mutation of the FH gene (c.422G>A, p.Trp141X). PMID: 22713448
  38. Tumor-derived FH and SDH mutations accumulate fumarate and succinate, leading to enzymatic inhibition of multiple alpha-KG-dependent dioxygenases and consequent alterations of genome-wide histone and DNA methylation. PMID: 22677546
  39. Inactivation of the TCA cycle enzyme, fumarate hydratase (FH), drives a metabolic shift to aerobic glycolysis in FH-deficient kidney tumors. PMID: 21907923
  40. The crystal structure of human fumarate hydratase shows that mutations can be grouped into two distinct classes: either affecting the structural integrity of the core enzyme architecture or localized around the enzyme active site. PMID: 21445611
  41. Reduced FH leads to the accumulation of hypoxia-inducible factor-2alpha (HIF-2alpha). PMID: 21695080
  42. Novel mutations within the FH gene are associated with hereditary leiomyomatosis and renal cell cancer. PMID: 21398687
  43. Four novel mutations and one whole-gene deletion of fumarate hydratase in families with an autosomal dominant syndrome characterized by multiple cutaneous piloleiomyomas, uterine leiomyomas, and papillary type 2 renal cancer. PMID: 20618355
  44. These experiments demonstrated that upregulation of HIF-1alpha occurs as a direct consequence of FH inactivation. PMID: 20660115
  45. This is the first case of fumaric aciduria described in Brazil, which presented with some interesting clinical and biochemical findings such as colpocephaly, hepatic alterations, and marked metabolic acidosis since birth. PMID: 20549362
  46. Data suggest that fumarase and fumaric acid are critical elements of the DNA damage response, which underlies the tumor suppressor role of fumarase in human cells and is most probably HIF independent. PMID: 20231875
  47. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata, and papillary renal cell cancer. PMID: 11865300
  48. Changes in the turnover number and the cocrystal structure with bound citrate due to the human missense mutation G-955-C engineered in fumarase C from Escherichia coli. PMID: 12021453
  49. Our findings indicate that mutations in fumarate hydratase do not play a major role in the development of sporadic leiomyosarcomas or uterine leiomyomas. PMID: 12177782
  50. Review: Fumarate hydratase catalyzes a step in the Krebs tricarboxylic-acid cycle. Inherited heterozygous mutations in the gene encoding this enzyme cause predisposition to inherited neoplasia syndromes. PMID: 12612654

Show More

Hide All

Database Links

HGNC: 3700

OMIM: 136850

KEGG: hsa:2271

STRING: 9606.ENSP00000355518

UniGene: Hs.592490

Involvement In Disease
Fumarase deficiency (FMRD); Hereditary leiomyomatosis and renal cell cancer (HLRCC)
Protein Families
Class-II fumarase/aspartase family, Fumarase subfamily
Subcellular Location
[Isoform Mitochondrial]: Mitochondrion.; [Isoform Cytoplasmic]: Cytoplasm, cytosol. Nucleus. Chromosome.
Tissue Specificity
Expressed in red blood cells; underexpressed in red blood cells (cytoplasm) of patients with hereditary non-spherocytic hemolytic anemia of unknown etiology.

Q&A

What are the primary monoclonal antibodies currently used in FH treatment and research?

The primary monoclonal antibodies (mAbs) used in Familial Hypercholesterolemia (FH) research and treatment target two key proteins involved in lipid metabolism: PCSK9 and ANGPTL3. The PCSK9 inhibitors include evolocumab and alirocumab, which have shown significant efficacy in reducing LDL-C levels, particularly in heterozygous FH (HeFH) patients . More recently, evinacumab, which targets ANGPTL3, has demonstrated promising results even in homozygous FH (HoFH) patients carrying null LDLR mutations . These mAbs represent different mechanisms of action, with PCSK9 inhibitors requiring residual LDLR activity, while ANGPTL3 inhibitors function through an LDLR-independent pathway.

How do PCSK9 monoclonal antibodies differ in their mechanism of action from other lipid-lowering therapies?

PCSK9 monoclonal antibodies function through a distinctive mechanism compared to conventional lipid-lowering therapies like statins:

  • Mechanism of action: PCSK9 inhibitors bind to the PCSK9 protein, preventing it from binding to low-density lipoprotein receptors (LDLRs). This prevents LDLR degradation, allowing more receptors to remain on the cell surface to clear LDL-C from circulation .

  • Dependence on LDLR functionality: Unlike statins which upregulate LDLR expression, PCSK9 inhibitors require the presence of functional LDLRs to be effective. This explains why patients with null LDLR mutations show limited or no response to PCSK9 inhibitors .

  • Complementary effects: When combined with statins, PCSK9 inhibitors provide additive benefits, as statins increase LDLR expression while PCSK9 inhibitors prolong LDLR lifespan on the cell surface, making them particularly valuable in FH patients who often require combination therapy .

What is the importance of LDLR mutation status in predicting response to monoclonal antibody therapy in FH?

LDLR mutation status is crucial for predicting therapeutic response to monoclonal antibody therapy in FH patients:

  • HeFH patients: Generally respond well to PCSK9 inhibitors due to having at least one functional LDLR allele, with LDL-C reductions typically ranging from 40-60% .

  • HoFH patients with defective mutations: Show variable responses to PCSK9 inhibitors based on residual LDLR activity. The TESLA part B trial demonstrated that patients with receptor defective mutations in one or both alleles achieved significant LDL-C reductions (40.8% compared to placebo) .

  • HoFH patients with null mutations: Exhibit minimal to no response to PCSK9 inhibitors due to absence of functional LDLRs. The TESLA trials showed that patients with null mutations in both LDLR alleles did not respond to evolocumab treatment .

  • Alternative approaches: For patients with null LDLR mutations, ANGPTL3 inhibitors like evinacumab offer promise through their LDLR-independent mechanism of action .

Understanding the specific LDLR mutation is therefore essential for therapeutic decision-making and for predicting treatment efficacy.

What methodological approaches are optimal for assessing antibody cooperativity in monoclonal antibody pairs targeting FH-related proteins?

Antibody cooperativity, where antibody pairs promote enhanced bactericidal killing compared to individual antibodies, requires sophisticated methodological approaches for assessment:

  • 3D electron microscopy: This technique allows for structural characterization of mAb-antigen-mAb cooperative complexes, revealing critical spatial arrangements. Research has shown that the angle formed between the antigen binding fragments (fAbs) assumes regular conformations that facilitate cooperativity .

  • In vitro binding assays: These assays determine simultaneous binding of cooperative mAb pairs and their target proteins. For example, studies with factor H-binding protein (fHbp) have demonstrated that certain mAb pairs can bind simultaneously and stably to both fHbp and human factor H (fH) in vitro .

  • Complement-mediated bactericidal activity testing: This approach assesses whether non-bactericidal mAbs in combination can elicit complement-mediated bactericidal activity. The JAR 41 mAb, for instance, demonstrated this capability when combined with other anti-fHbp mAbs .

  • Epitope mapping: Detailed epitope mapping is essential to identify binding sites that allow for cooperative interactions. Techniques include X-ray crystallography, hydrogen-deuterium exchange mass spectrometry, and mutagenesis studies to precisely locate epitopes that permit simultaneous binding of multiple antibodies .

  • In vivo models: Human factor H transgenic rat models have been used to evaluate passive protection against bacteremia, providing a relevant system to validate cooperativity observed in vitro .

How do we reconcile the heterogeneity in lipid profile responses to PCSK9 antibody therapy across different FH genotypes?

The heterogeneity in lipid profile responses to PCSK9 antibody therapy across FH genotypes can be reconciled through several analytical approaches:

  • Genotype-stratified analysis: Data should be stratified based on specific LDLR mutation types (null vs. defective) and zygosity (heterozygous vs. homozygous). Evidence from clinical trials shows significantly different responses, with LDLR-negative HoFH patients showing minimal response compared to LDLR-defective patients .

  • Residual LDLR function quantification: Developing standardized assays to quantify residual LDLR activity could help predict response magnitude. The TESLA trials demonstrated variable responses even among patients with the same LDLR mutation, with reductions ranging from 7.1% to 56.0% .

  • Meta-regression techniques: Pooled analyses incorporating LDLR functionality as a continuous variable rather than a categorical one may better explain response variability. One meta-analysis found that heterogeneity in lipid profile analyses was partly caused by the different types of FH (HoFH or HeFH) .

  • Consideration of modifying factors: Other genetic modifiers, environmental factors, and baseline lipid profiles should be incorporated into prediction models. For example, patients with extremely high baseline LDL-C levels may show greater absolute but lower percentage reductions .

  • Longitudinal response patterns: Analysis of lipid trajectories over time rather than single timepoint measurements provides more comprehensive understanding of therapeutic efficacy across genotypes.

What are the optimal experimental designs for evaluating monoclonal antibody charge variants in the context of FH research?

Optimal experimental designs for evaluating monoclonal antibody charge variants in FH research require sophisticated approaches:

  • Refined calibration in imaged capillary Iso Electric Focusing (icIEF): Recent advances demonstrate that refining calibration approaches in icIEF methods allows for obtaining more reliable and objective isoelectric points (pIs), providing deeper understanding of pH gradients along capillaries .

  • "Unbiased" Experimental Design (UED): This approach minimizes bias by studying resolution as a multivariate function of different input variables, enabling development of optimal methods tailored to specific pH ranges .

  • Charge Variants Profile Assessment (CVPA): Rather than relying on relative comparisons with reference standards, developing objective parameters for charge variant profiles helps overcome inconsistent outputs across different instruments .

  • Multiparameter monitoring: Simultaneously tracking multiple post-translational modifications (PTMs) such as glycosylation and deamidation that may affect drug efficacy and safety in FH treatment .

  • Stability-indicating methods: Incorporating stress conditions (temperature, pH, oxidation) to predict charge variant formation during manufacturing and storage, which is particularly relevant for maintaining consistent efficacy in long-term FH treatment regimens.

What are the key considerations for designing clinical trials to evaluate monoclonal antibody efficacy in FH patients with varying genetic backgrounds?

Designing clinical trials to evaluate monoclonal antibody efficacy in genetically diverse FH populations requires meticulous planning:

  • Genetic stratification: Trials should prospectively stratify participants based on:

    • LDLR mutation status (null vs. defective)

    • Zygosity (HeFH vs. HoFH)

    • Other causative genes (APOB, PCSK9, LDLRAP1)

    This stratification is crucial as response rates vary significantly between these groups.

  • Sample size considerations: Adequate powering for subgroup analyses is essential:

    FH TypeMinimum Subjects NeededExpected LDL-C Reduction
    HeFH40-60 per arm40-60%
    HoFH (defective)15-20 per arm20-40%
    HoFH (null)20-25 per arm0-10% (PCSK9 inhibitors)
    30-50% (ANGPTL3 inhibitors)
  • Endpoint selection: Beyond traditional LDL-C reduction, consider:

    • Effects on other lipoproteins (Lp(a), VLDL)

    • Cardiovascular outcomes

    • Quality of life measures

    • Long-term safety profiles

  • Control arm design: For rare HoFH populations, consider:

    • Crossover designs

    • Intra-patient controlled studies

    • Historical control comparisons

    • Adaptive trial designs

  • Biomarker development: Incorporate exploratory biomarkers to identify predictors of response and guide personalized treatment approaches.

  • Duration considerations: Include extended follow-up periods to assess long-term efficacy and safety, particularly important for lifelong conditions like FH.

How can researchers optimize monoclonal antibody epitope mapping for broadly cross-reactive antibodies against FH-relevant targets?

Optimizing epitope mapping for broadly cross-reactive monoclonal antibodies against FH-relevant targets requires sophisticated approaches:

  • Combined structural techniques: Integrating X-ray crystallography with cryo-electron microscopy provides comprehensive structural insights. This approach has revealed, for example, that the JAR 41 mAb epitope is located on a conserved region of the N-terminal portion of the fHbp molecule opposite that of fH contact residues, explaining its broad cross-reactivity .

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS): This technique measures the rate of hydrogen/deuterium exchange in protein backbones, identifying protected regions upon antibody binding. HDX-MS is particularly valuable for conformational epitope mapping when crystallization is challenging.

  • Alanine scanning mutagenesis: Systematic replacement of amino acids with alanine helps identify critical binding residues. Research on factor H binding protein (fHbp) utilized this approach to map epitopes for broadly cross-reactive antibodies like JAR 41 .

  • Competitive binding assays: These assays determine whether antibodies recognize overlapping or non-overlapping epitopes, essential for identifying antibodies that can bind simultaneously to enhance efficacy.

  • Cross-variant binding studies: Systematically testing antibody binding across protein variants from all groups (e.g., all three fHbp variant groups) to confirm true broad cross-reactivity .

  • Functional correlation studies: Correlating epitope location with functional outcomes, such as complement-mediated bactericidal activity or inhibition of PCSK9/LDLR interaction, to identify the most therapeutically relevant epitopes.

What are the optimal protocols for assessing PCSK9 inhibitor efficacy in preclinical FH models?

Optimal protocols for assessing PCSK9 inhibitor efficacy in preclinical FH models include:

  • Selection of appropriate animal models:

    • LDLR-deficient mice (mimicking HoFH)

    • LDLR-defective mice (mimicking HeFH)

    • Human LDLR transgenic mice with specific mutations

    • PCSK9 transgenic mice

    • Humanized liver chimeric mice expressing human LDLR variants

  • Rigorous study design elements:

    • Randomization procedures

    • Blinded assessment

    • Appropriate control groups

    • Sample size calculation based on expected effect sizes

    • Longitudinal assessment points

  • Comprehensive lipid profiling:

    • Frequent sampling timeline (baseline, 24h, 48h, 7d, 14d, 28d)

    • Complete lipoprotein analysis (LDL-C, HDL-C, total cholesterol, triglycerides)

    • Lipoprotein particle number and size assessment

    • ApoB and ApoA1 quantification

    • Lp(a) measurement in humanized models

  • Pharmacokinetic/pharmacodynamic (PK/PD) assessment:

    • Antibody concentration measurements

    • PCSK9 level monitoring (free vs. bound)

    • Correlation between PCSK9 suppression and LDL-C reduction

    • Dosing frequency optimization

  • Mechanistic assessments:

    • LDLR expression quantification (protein and mRNA levels)

    • Hepatic LDL clearance rates

    • De novo cholesterol synthesis rates

    • VLDL production rates

What analytical techniques are most reliable for characterizing monoclonal antibody heterogeneity in FH therapeutic applications?

For characterizing monoclonal antibody heterogeneity in FH therapeutic applications, several analytical techniques have proven particularly reliable:

  • Charge variant analysis:

    • Imaged capillary Iso Electric Focusing (icIEF): Provides high-resolution separation of charge variants based on isoelectric point (pI)

    • Cation exchange chromatography (CEX): Complements icIEF for charge variant profiling

    • Innovative calibration approaches to ensure consistent pI determination across different instruments and laboratories

  • Size variant analysis:

    • Size exclusion chromatography (SEC): Detects aggregates, fragments and monomers

    • Analytical ultracentrifugation (AUC): Provides information on size distribution and conformation

    • Dynamic light scattering (DLS): Rapid assessment of size distribution

  • Structural characterization:

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS): Reveals conformational dynamics

    • Differential scanning calorimetry (DSC): Assesses thermal stability

    • Circular dichroism (CD): Monitors secondary structure

  • Post-translational modification analysis:

    • Liquid chromatography-mass spectrometry (LC-MS): Identifies and quantifies PTMs including glycosylation, deamidation, and oxidation

    • Capillary electrophoresis with laser-induced fluorescence (CE-LIF): Analyzes glycan profiles

    • Site-specific glycopeptide mapping: Determines glycan microheterogeneity

  • Functional assessment:

    • Surface plasmon resonance (SPR): Measures binding kinetics

    • Cell-based potency assays: Correlates analytical attributes with biological function

    • Fc receptor binding assays: Assesses effector function capability

How can researchers effectively design combination studies of different monoclonal antibody classes for treating resistant forms of FH?

Designing effective combination studies of different monoclonal antibody classes for resistant forms of FH requires a systematic approach:

  • Rational target selection:

    • Combine antibodies targeting complementary pathways (e.g., PCSK9 inhibitors plus ANGPTL3 inhibitors)

    • Focus on mechanisms that work independently of LDLR function for null-mutation HoFH patients

    • Consider targeting multiple steps in the same pathway for synergistic effects

  • In vitro screening approach:

    Study PhaseMethodologyExpected Outcome
    Initial ScreeningHepatocyte cultures from FH patientsIdentify promising combinations
    Mechanism ValidationReceptor binding competition assaysConfirm non-competitive binding
    Dose-Response AssessmentCheckerboard titration in FH cell modelsDetermine optimal dose ratios
  • Preclinical design elements:

    • Use multiple animal models representing different FH genotypes

    • Test sequential vs. simultaneous administration

    • Evaluate different dosing intervals and ratios

    • Monitor for unexpected antagonistic effects

  • Clinical trial design considerations:

    • Adaptive trial designs with interim analyses

    • Factorial study designs to assess contributions of each component

    • Crossover elements where feasible

    • Rich PK/PD sampling to understand interactions

    • Safety monitoring committees with expertise in immunogenicity

  • Specialized outcome measures:

    • Combined efficacy metrics beyond LDL-C (e.g., comprehensive lipoprotein profiling)

    • Assessment of cardiovascular imaging biomarkers

    • Quality of life and treatment burden measures

    • Long-term monitoring for novel toxicities

What standardized approaches should be adopted for evaluating immunogenicity of monoclonal antibodies in long-term FH management?

Standardized approaches for evaluating immunogenicity of monoclonal antibodies in long-term FH management should include:

  • Tiered testing strategy:

    • Screening assay: Highly sensitive electrochemiluminescence or ELISA to detect total anti-drug antibodies (ADA)

    • Confirmation assay: Competitive inhibition to confirm specificity

    • Characterization assays: Determine if ADAs are neutralizing

    • Titer determination: Semi-quantitative measurement of ADA levels

  • Sampling schedule optimization:

    • Baseline (pre-dose) sampling to detect pre-existing antibodies

    • Early phase sampling (weeks 4-12) to capture primary immune responses

    • Long-term sampling (6-12 months and annually thereafter) to monitor persistence

    • Additional samples when efficacy wanes or adverse events occur

  • Clinical correlation assessment:

    • Relation between ADA development and efficacy parameters (LDL-C changes)

    • Pharmacokinetic impact analysis (drug clearance rates)

    • Hypersensitivity and injection site reaction correlation

    • Cross-reactivity with endogenous proteins (PCSK9, ANGPTL3)

  • Risk-based considerations for special populations:

    • HoFH patients requiring lifelong therapy

    • Pediatric FH patients with developing immune systems

    • Patients switching between different monoclonal antibodies

    • Patients on immunomodulatory medications

  • Standardized reporting parameters:

    • Incidence of treatment-emergent ADAs

    • ADA persistence duration

    • Neutralizing antibody frequency

    • Clinical impact categorization (none, reduced efficacy, loss of efficacy, adverse events)

What are the emerging approaches for developing next-generation monoclonal antibodies that overcome current limitations in FH therapy?

Emerging approaches for next-generation monoclonal antibodies in FH therapy include:

  • Bispecific antibody development:

    • PCSK9/ANGPTL3 bispecific antibodies to simultaneously target multiple lipid metabolism pathways

    • LDLR/PCSK9 bispecifics to enhance receptor upregulation and prevention of degradation

    • Design of antibodies that can function independently of LDLR status for HoFH patients with null mutations

  • Extended half-life technologies:

    • Fc engineering to enhance neonatal Fc receptor (FcRn) binding

    • Albumin fusion constructs

    • Polyethylene glycol (PEG) conjugation
      These approaches could potentially reduce dosing frequency from biweekly/monthly to quarterly administration

  • Tissue-specific targeting:

    • Hepatocyte-targeted antibody constructs to increase liver specificity

    • Reduced systemic exposure to minimize off-target effects

    • Enhanced penetration into hepatic tissue

  • Alternative administration routes:

    • Subcutaneous formulations with enhanced stability and concentration

    • Oral delivery systems using antibody fragments

    • Novel device-based delivery methods

  • Combination therapy optimization:

    • Fixed-dose combinations with small molecule inhibitors

    • Synergistic antibody cocktails targeting multiple pathways

    • Personalized combination approaches based on genetic profile

How might artificial intelligence and machine learning accelerate epitope discovery for broadly effective monoclonal antibodies in FH?

Artificial intelligence and machine learning approaches are poised to revolutionize epitope discovery for broadly effective monoclonal antibodies in FH through several mechanisms:

  • Predictive epitope mapping:

    • Deep learning models trained on known antibody-antigen crystal structures can predict epitopes on novel targets

    • Natural language processing of scientific literature to identify potential epitope regions not previously considered

    • Integration of sequence conservation, structural features, and physicochemical properties to rank epitope candidates

  • Antibody design optimization:

    • Generative adversarial networks (GANs) to design novel antibody sequences targeting specific epitopes

    • Reinforcement learning to optimize antibody properties (affinity, specificity, developability)

    • Neural networks to predict antibody-antigen binding affinity from sequence alone

  • Virtual screening approaches:

    • Molecular dynamics simulations to identify stable binding conformations

    • Graph neural networks for predicting protein-protein interactions

    • Quantum mechanical calculations to estimate binding energies

  • Cross-reactivity prediction:

    • Machine learning models to predict antibody binding across variant proteins (e.g., PCSK9 variants)

    • Algorithms to identify conserved structural epitopes despite sequence variation

    • Models to distinguish binding from functional neutralization

  • Clinical response prediction:

    • Integration of patient genetic data with antibody binding properties to predict response

    • Systems biology approaches to model antibody effects on lipid metabolism networks

    • Real-world evidence analysis to identify patterns in treatment outcomes

What are the best practices for translating preclinical findings on monoclonal antibody efficacy to clinical applications in FH?

Translating preclinical findings on monoclonal antibody efficacy to clinical applications in FH requires adherence to several best practices:

  • Robust animal model selection:

    • Use models that accurately recapitulate the specific genetic defects found in FH patients

    • Employ humanized models expressing human LDLR variants when possible

    • Consider using patient-derived hepatocytes for in vitro validation

  • Comprehensive pharmacological assessment:

    • Establish clear exposure-response relationships

    • Determine minimum effective concentrations

    • Characterize target engagement metrics

    • Assess off-target binding potential

  • Allometric scaling considerations:

    • Apply species-specific correction factors for antibody clearance

    • Account for differences in target expression levels

    • Consider target turnover rates when predicting human dosing

  • Biomarker qualification and validation:

    • Identify translational biomarkers that function across species

    • Validate surrogate endpoints (e.g., LDL-C reduction) against clinical outcomes

    • Develop assays with equivalent performance in preclinical and clinical settings

  • Patient stratification strategy development:

    • Define genetic and phenotypic factors that predict response

    • Create algorithms for patient selection based on preclinical response patterns

    • Develop companion diagnostics when appropriate

What are the key methodological considerations for conducting real-world evidence studies on monoclonal antibody effectiveness in diverse FH populations?

Conducting real-world evidence studies on monoclonal antibody effectiveness in diverse FH populations requires careful methodological planning:

  • Study design selection:

    • Prospective registries with standardized data collection

    • Electronic health record-based cohort studies

    • Nested case-control studies for rare outcomes

    • Pragmatic clinical trials with minimal exclusion criteria

  • Population representativeness:

    • Include underrepresented populations often excluded from randomized trials

    • Capture various FH genotypes, including rare mutations

    • Include pediatric and elderly populations

    • Consider geographic and ethnic diversity to capture genetic variation

  • Outcome standardization:

    • Develop consistent definitions for effectiveness (e.g., percent LDL-C reduction, achievement of target levels)

    • Standardize cardiovascular outcome assessments

    • Create uniform adverse event recording protocols

    • Establish quality of life and adherence metrics

  • Confounding management:

    • Apply propensity score methods to balance treatment groups

    • Use instrumental variable analyses where appropriate

    • Implement marginal structural models for time-varying confounding

    • Conduct sensitivity analyses for unmeasured confounders

  • Data quality assurance:

    • Implement source data verification procedures

    • Develop algorithms to identify missing or implausible values

    • Create data completeness metrics

    • Establish standards for genetic testing quality

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 2025 TheBiotek. All Rights Reserved.