TF Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Overview of TF Monoclonal Antibody

Tissue Factor (TF) Monoclonal Antibodies are laboratory-engineered immunoglobulins designed to specifically bind to tissue factor, a transmembrane glycoprotein critical in initiating blood coagulation and implicated in cancer progression . These antibodies are produced by cloning white blood cells to target TF’s extracellular domain, enabling precise therapeutic and diagnostic applications . TF is overexpressed in malignancies such as glioblastoma, pancreatic cancer, and ovarian adenocarcinoma, making it a key biomarker for targeted therapies .

Mechanism of Action

TF monoclonal antibodies exert their effects through multiple pathways:

  • Direct Tumor Targeting: Bind to TF on cancer cells, blocking procoagulant activity and TF-FVIIa signaling pathways involved in tumor growth and metastasis .

  • Antibody-Drug Conjugates (ADCs): Deliver cytotoxic agents (e.g., monomethyl auristatin E/MMAE) directly to TF-expressing cells, inducing apoptosis .

  • Immune Activation: Engage Fc-mediated effector functions like antibody-dependent cellular cytotoxicity (ADCC) to eliminate tumor cells .

  • Coagulation Inhibition: Neutralize TF’s interaction with clotting factors (e.g., Factor X), reducing thrombotic risks in cancer patients .

Preclinical Studies

ApplicationModelOutcomeSource
Glioblastoma ImagingMouse intracranial tumorsAnti-TF mAb (clone 1849) enabled fluorescence and SPECT/CT imaging of tumors with 98% specificity .
Pancreatic CancerPatient-derived xenograftsADC (ALT836 conjugated with ⁸⁶/⁹⁰Y) showed tumor uptake correlated with TF expression levels .
Ovarian CancerSK-OV-3 xenograftsTF-011-MMAE ADC induced complete tumor regression in 80% of cases .

Clinical Trials

  • Tisotumab Vedotin: An ADC targeting TF demonstrated 24% objective response rate in recurrent cervical cancer .

  • Thrombosis Management: Anti-TF mAb (AP-1) reduced reocclusion rates by 60% in rabbit carotid artery models post-thrombolysis .

Cancer Treatment

  • Solid Tumors: TF-targeted ADCs like TF-011-MMAE achieved complete regression in heterogeneous tumors, even with low TF expression .

  • Glioblastoma: Anti-TF mAbs aid in surgical guidance by highlighting necrotic tumor regions .

Anticoagulation Therapy

  • Factor X Inhibition: Monoclonal antibody TF4A12 prolonged prothrombin time by 300% in vitro, offering potential for treating thrombosis without bleeding risks .

Diagnostic Uses

  • Immuno-SPECT Imaging: ¹¹¹In-labelled anti-TF mAbs provide high-resolution imaging for glioma grading and biopsy targeting .

  • Histopathology: Clone 1849 antibodies identify necrosis and pseudopalisading cells in glioblastoma specimens .

Challenges and Limitations

  • Heterogeneous TF Expression: Requires combination therapies for tumors with variable antigen density .

  • Immune Reactions: Early murine antibodies (e.g., AP-1) faced immunogenicity issues, mitigated by humanized designs .

  • Drug Resistance: Observed in 15% of ADC-treated patients due to TF downregulation .

Future Directions

  • Bispecific Antibodies: Combining TF targeting with immune checkpoint inhibitors (e.g., anti-PD-1) .

  • Next-Gen ADCs: Utilizing non-cleavable linkers (e.g., mc-MMAF) to improve tumor selectivity .

Product Specs

Buffer
PBS, pH 7.4, containing 0.02% sodium azide as preservative and 50% Glycerol.
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days after receiving them. Delivery time may vary based on the purchase method or location. Please consult your local distributors for specific delivery information.
Synonyms
Apotransferrin antibody; Beta 1 metal binding globulin antibody; Beta-1 metal-binding globulin antibody; DKFZp781D0156 antibody; PRO1400 antibody; PRO1557 antibody; PRO2086 antibody; Serotransferrin antibody; Serotransferrin precursor antibody; Siderophilin antibody; TF antibody; TFQTL1 antibody; Transferin antibody; Transferrin antibody; TRFE_HUMAN antibody
Target Names
TF
Uniprot No.

Target Background

Function
Transferrins are iron-binding transport proteins capable of binding two Fe(3+) ions in conjunction with the binding of an anion, usually bicarbonate. They play a crucial role in transporting iron from absorption and heme degradation sites to storage and utilization sites. Serum transferrin may also contribute to cell proliferation stimulation. In microbial infections, transferrin serves as an iron source for Neisseria species, which capture the protein and extract its iron for their own use.
Gene References Into Functions
  • High transferrin expression is linked to Spontaneous intracranial hypotension. PMID: 29621631
  • SNPs rs4525863, rs1880669, and rs2692695 within the TF gene might act as a protective factor against ischemic stroke in the Southern Chinese Han population. PMID: 29936663
  • Our research confirms the critical role of blood TSAT in experimental stroke-induced brain damage. Furthermore, our findings suggest that the protective effect of iron-free transferrin (apotransferrin, ATf) at the neuronal level involves preventing NMDA-induced HTf uptake and ROS production, ultimately reducing neuronal damage. PMID: 29248829
  • Histochemical staining using anti-Tf antibody and an N-acetylglucosamine-binding lectin suggests that brain-type Tf is secreted from the choroid plexus, the tissue responsible for CSF production. PMID: 29701803
  • Data indicate a positive association between serum levels of iron status biomarkers (ferritin, transferrin, and hemoglobin) and the risk of hyperuricemia in Chinese adults. PMID: 29425155
  • Transferrin saturation was associated with less severe anemia in early CKD patients. PMID: 29227972
  • Three single nucleotide polymorphisms associated with iron regulation were genotyped in multiple sclerosis: two in the human hereditary hemochromatosis protein gene HFE: rs1800562 (C282Y mutation) and rs1799945 (H63D mutation), and rs1049296 SNP in the transferrin gene (C2 mutation). We observed a higher prevalence of TF-C2 in multiple sclerosis patients. PMID: 29201641
  • Increased serum transferrin and ceruloplasmin turnover have been reported in diet-controlled patients with type 2 diabetes. PMID: 29079528
  • The model predicts that small vesicles promote activation of FX by the extrinsic tenase (VIIa/TF) significantly better than large vesicles. PMID: 28935233
  • This work presents an HPLC analysis of carbohydrate-deficient transferrin. PMID: 28971232
  • Transferrin, associated with Adenoid Cystic Carcinoma of the Parotid Gland in an immunohistochemical study, was found to be up-regulated in Pleomorphic Adenoma of the Parotid Gland. PMID: 28497265
  • The RNAi knock-down of Trypanosoma brucei GSK3beta reduced Tf endocytosis. PMID: 27626104
  • Holotransferrin treatment to recombinant-TfR1 HepG2 cells did not elevate HAMP responses compared to untreated or wild-type cells. PMID: 27667164
  • Blood lead levels showed a positive association with plasma homocysteine levels measured 4-6 years later, and three SNPs in the TF gene modified the association. PMID: 28775131
  • This technical note presents the aberrant glycosylation profiles of transferrin glycoforms in congenital disorder of glycosylation patients to shed light on the Mass Spectrometry of native transferrin and glycopeptides from a clinical glycoproteomics perspective. PMID: 27095603
  • Over the first 500 s, approximately 92,000 molecules of thrombin were generated per surface TF molecule for the 250-mum-long coating. PMID: 27605669
  • High carbohydrate-deficient transferrin is associated with relapse in alcohol-dependent patients. PMID: 26851349
  • High Plasma Carbohydrate-deficient transferrin is associated with monoclonal light chain gammopathy. PMID: 27003373
  • The value of transferrin receptors (TfRs)/cell reveals a 100-fold increase in the number of TfRs per bEnd3 cells compared to human umbilical vein cells. PMID: 27100851
  • During pregnancy, the levels of 2-, 5- and 6-sialo transferrin isoforms increased, while 3- and 4-sialo transferrin isoforms decreased. PMID: 27448041
  • Alterations in transferrin glycosylation/sialylation in pancreatic cancer differ from those observed in some other cancers. PMID: 27312554
  • In a multiple regression analysis, FN3K rs1056534, TF polymorphism, and the presence of diabetes mellitus were predictors for HHV-8 infection. PMID: 27461879
  • The analysis of the hTf complex with a bacterial receptor that has evolved to sequester iron identifies two regions contacting rapidly evolving residues that mechanically manipulate dissociation from the pathogen. PMID: 26955866
  • Transferrin mutations can significantly impair the diagnostics of chronic alcohol abuse. PMID: 26333807
  • Total iron binding capacity was strongly associated with variants in and near the TF gene, and with variants in the HFE gene in premenopausal women of European descent. PMID: 26852655
  • Data suggest that up-regulation of serum transferrin levels is positively associated with the risk of metabolic syndrome and overweight/obesity, but not with the risk of diabetes among Chinese men and women. PMID: 25914390
  • Activation of PI3K/Akt by retinoic acid modulates AMPK activity in ECs and plays a crucial role in the inhibition of coagulatory factors such as TF, PAI-1, and HMGB1 in inflammatory conditions. PMID: 26116962
  • TF served as the carrier to deliver irons and could directly stimulate cardiomyocytes hypertrophy. PMID: 26099594
  • Data show that transferrin (Tf) was attached to nanoparticles with an acid-cleavable linkage that facilitates the release of nanoparticles from Tf that are bound to Tf receptors (TfR) during transcytosis, promoting entry of the nanoparticles into the brain. PMID: 26392563
  • H2O2 induces the expression of transferrin, and consequently, decreased iron absorption, suggesting a novel mechanism for iron deficiency in pediatric non-alcoholic steatohepatitis patients. PMID: 25000850
  • Human transferrin is internalized across the mycobacterial cell wall in a GAPDH-dependent manner within infected macrophages. PMID: 25163484
  • Data suggest that the 15 disulfide bonds in transferrin vary in their susceptibility to reduction and their contribution to protein conformation/stability. PMID: 25716754
  • Apelin-13 has a role in inducing expression of prothrombotic tissue factor. PMID: 25298206
  • The rs3811647 polymorphism in the TF gene was identified as the only SNP significantly associated with iron metabolism through serum transferrin and iron levels. PMID: 25457201
  • Significant associations between serum iron binding capacity levels and two SNPs around TF on chromosome 3 were identified. PMID: 25224454
  • Hereditary hypotransferrinemia caused by novel transferrin mutations can lead to elevated transferrin saturation and, when associated with HFE or HAMP mutations, to iron overload. PMID: 25486930
  • Results show that higher TF expression in patients with hypereosinophilic disorders may contribute to an increased thrombotic risk. PMID: 25375118
  • Genetic association studies in a population of black women in South Africa: Data confirm that an SNP in TF (rs1799852) is associated with iron status/iron-deficiency anemia in the studied population. PMID: 25809685
  • Decreased circulating levels in abdominal aortic aneurysm. PMID: 24599423
  • Infants born to mothers with the HFE C282Y gene missense variant but not the TF P570S or HFE H63D gene missense variants had lower umbilical cord blood lead levels compared to those born to women who were wild-type. PMID: 25287020
  • TF translocates to the cell front in association with cytoskeleton proteins and regulates HVSMC migration by mechanisms dependent and independent of factor (F)VIIa/PAR2. PMID: 22938499
  • No association was found between the c.-2G>A polymorphism and keratoconus; no association was found between transferrin polymorphisms and Fuchs endothelial corneal dystrophy occurrence. PMID: 24350254
  • Understanding the interaction between [Cr(phen)3](3+) with transferrin is relevant because this protein could serve as a delivery agent of Cr(III) complex to tumor cells. PMID: 24972167
  • Increased urinary excretion of plasma proteins such as IgG, ceruloplasmin, and transferrin, with different molecular radii of 55 A or less and different isoelectric points, precedes the development of microalbuminuria in patients with NIDDM and hypertension. PMID: 24256706
  • Elevated BMI reduces the diagnostic utility of carbohydrate-deficient transferrin at higher alcohol intake in subjects with liver disease. PMID: 23875541
  • Data suggest that there is no association between the plasma level of carbohydrate-deficient transferrin and volume decrease of brain gray or white matter in male subjects as a result of current/recent or lifetime alcohol drinking in the Netherlands. PMID: 23027680
  • Studies indicate that several genes have been linked to iron homeostasis, including transferrin (TF), iron regulatory protein 1 (ACO1), and transferrin receptor 2 (TFR2). PMID: 23817740
  • The A allele of SNP rs3811647 increases Tf expression, which might underlie inter-individual variation in serum transferrin levels. PMID: 23588470
  • Transferrin mutations were found in several members of two non-consanguineous families with hypochromic microcytic anemia & hemodiserosis. The missense mutations were 1940 A>T & 1825 C>T. PMID: 23888904
  • The structures of hTF observed here represent key conformers captured in the dynamic nature of the transferrin family proteins and provide a structural basis for understanding the mechanism of metal uptake and release in transferrin families. PMID: 23256035

Show More

Hide All

Database Links

HGNC: 11740

OMIM: 190000

KEGG: hsa:7018

STRING: 9606.ENSP00000385834

UniGene: Hs.518267

Involvement In Disease
Atransferrinemia (ATRAF)
Protein Families
Transferrin family
Subcellular Location
Secreted.
Tissue Specificity
Expressed by the liver and secreted in plasma.

Q&A

What is Tissue Factor (TF) and why are monoclonal antibodies against it scientifically significant?

Tissue Factor (TF), also known as coagulation Factor III, is a cell surface glycoprotein that serves as the primary initiator of the blood coagulation cascade. It functions as a high-affinity receptor for coagulation Factor VII, which when activated forms a complex that triggers downstream coagulation factors, ultimately leading to fibrin clot formation and platelet aggregation .

Monoclonal antibodies targeting TF have emerged as powerful tools for multiple scientific applications:

  • Hemostasis and thrombosis research: TF has been implicated as a critical controlling molecule in hemostasis, thrombosis, and inflammation, making anti-TF antibodies valuable for studying these processes .

  • Cancer biology: TF is overexpressed in various malignancies, with expression levels often correlating with tumor grade. In gliomas, TF expression increases proportionally with malignancy grade according to WHO classification, with remarkable expression in necrosis and pseudopalisading cells (hallmarks of glioblastoma multiforme) .

  • Diagnostic applications: Anti-TF monoclonal antibodies can visualize TF-expressing tumors, potentially aiding in diagnosis and biopsy guidance .

  • Therapeutic development: As anticoagulants, anti-TF antibodies can inhibit thrombosis with reduced systemic effects compared to conventional anticoagulants .

Anti-TF monoclonal antibodies provide researchers with specific molecular probes to investigate TF biology across these diverse areas, offering insights into both physiological and pathological processes.

How are TF monoclonal antibodies generated and what methodological approaches yield the most effective antibodies?

The generation of effective TF monoclonal antibodies involves several methodological approaches:

Antigen design strategies:

  • Whole recombinant TF protein (extracellular domain sTF1-219)

  • Strategically designed TF peptides (TFP) targeting specific functional regions, such as the Factor X binding site

  • Multiple antigenic peptides (TF-MAP) consisting of TF peptides coupled to polylysine matrix for enhanced immunogenicity

Immunization and production methods:

  • Traditional hybridoma technology using mouse immunization followed by B-cell fusion with myeloma cells

  • Repetitive immunizations at multiple sites (RIMMS) protocol, which can generate antibodies within a relatively short timeframe (5-6 weeks)

  • Phage display technology for in vitro antibody selection

  • Single B cell cloning for direct isolation of antibody-producing cells

  • Transgenic animals expressing human antibody repertoires

Screening and selection protocols:

  • Primary screening by ELISA against recombinant TF or TF peptides

  • Secondary validation using flow cytometry with TF-expressing cell lines

  • Functional assays such as coagulation inhibition tests (dilute prothrombin time)

  • Factor X activation assays to determine mechanism of action

  • Binding kinetics analysis using surface plasmon resonance (BIAcore)

Effective workflow example:
In one documented approach, researchers designed a TF peptide specific for the FX binding site, coupled it to a polylysine matrix as a multiple antigenic peptide (TF-MAP), immunized Balb/c mice, and generated hybridomas. This yielded the TF4A12 antibody with high anticoagulant potency due to its ability to specifically block FX binding to the TF/FVIIa complex without affecting FVII binding to TF .

What are the optimal detection methods for employing TF monoclonal antibodies in experimental systems?

TF monoclonal antibodies can be utilized across multiple detection platforms, each offering distinct advantages depending on research objectives:

Immunohistochemistry (IHC):

  • Methodology: Paraffin or frozen tissue sections treated with anti-TF antibodies followed by appropriate detection systems

  • Applications: Visualization of TF expression patterns in tumor samples; correlation with histopathological features

  • Example: Anti-TF 1849 mAb successfully demonstrated that TF expression in gliomas increases proportionally with malignancy grade according to WHO classification

Immunofluorescence (IF):

  • Methodology: Cell or tissue samples labeled with fluorophore-conjugated anti-TF antibodies

  • Applications: High-resolution subcellular localization of TF; co-localization studies with other markers

  • Available formats: Multiple conjugates including FITC, PE, and various Alexa Fluor® dyes

Flow cytometry:

  • Methodology: Single-cell suspensions labeled with fluorophore-conjugated anti-TF antibodies

  • Applications: Quantitative analysis of cell surface TF expression; sorting of TF-expressing cell populations

  • Example: Used to validate binding of anti-TF mAbs to native TF on stable cell lines expressing human TF

Western blotting:

  • Methodology: Protein extracts separated by SDS-PAGE and probed with anti-TF antibodies

  • Applications: Detection of TF protein expression levels; identification of different TF forms

  • Technical considerations: Most effective with antibodies recognizing linear epitopes

ELISA:

  • Methodology: Plate-bound antigen detected with anti-TF antibodies or sandwich formats

  • Applications: Quantification of TF in biological fluids; high-throughput screening

  • Sensitivity: Can detect picogram levels of TF with optimized protocols

Molecular imaging:

  • Methodology: Radiolabeled (e.g., 111In) or fluorophore-labeled anti-TF antibodies administered in vivo

  • Applications: Non-invasive visualization of TF-expressing tumors; monitoring treatment response

  • Example: Both fluorescence and SPECT/CT imaging using anti-TF 1849 IgG showed efficient accumulation in TF-overexpressing intracranial tumors in mice

Combinatorial approaches:
For comprehensive analysis, researchers often employ multiple detection methods. For instance, validation of a novel anti-TF antibody might begin with ELISA screening, followed by flow cytometry to confirm binding to native TF, immunohistochemistry to evaluate tissue distribution patterns, and functional assays to determine effects on coagulation.

How does the specificity of TF monoclonal antibodies affect their research applications and what validation methods should be employed?

The specificity of TF monoclonal antibodies significantly impacts their research utility and requires rigorous validation:

Determinants of specificity:

  • Epitope recognition (linear vs. conformational)

  • Cross-reactivity with related proteins

  • Species cross-reactivity (human, mouse, rat, etc.)

  • Recognition of different TF forms (full-length vs. alternatively spliced variants)

  • Ability to distinguish between encrypted (inactive) vs. decrypted (active) TF

Validation methodologies:

Validation MethodExperimental ApproachKey Parameters
Epitope mappingPeptide arrays, hydrogen-deuterium exchange, X-ray crystallographyPrecise epitope identification, structural data
Cross-reactivity testingELISA, Western blot with related proteinsSpecificity ratio, off-target binding
Species cross-reactivityTesting against TF from multiple speciesConservation of binding across species
Knockout/knockdown controlsTesting in TF-knockout cells/tissuesSignal absence in TF-negative samples
Functional validationCoagulation assays (PT, FX activation)Inhibitory potency, mechanism of action

Research application considerations:

For imaging applications, antibodies should:

  • Bind TF with high specificity and affinity (0.5-2 nM range)

  • Ideally not inhibit coagulation (to avoid systemic effects)

  • Have appropriate pharmacokinetics for the imaging modality

  • Target epitopes abundantly expressed on tumors

For functional studies, researchers should select antibodies that:

  • Target specific functional domains (e.g., FVII binding site, FX interaction region)

  • Have well-characterized effects on TF activity

  • Can distinguish between different functional states of TF

For quantitative analysis, validation should include:

  • Standard curves with recombinant TF

  • Recovery experiments in relevant biological matrices

  • Comparison with orthogonal detection methods

Case study in specificity validation:
Researchers developed a panel of murine monoclonal antibodies against human TF using a modified RIMMS protocol. Following primary ELISA screening, they validated specificity through:

  • Flow cytometry using a stable cell line expressing human TF

  • Blood coagulation assays to assess functional effects

  • Surface plasmon resonance (BIAcore) to determine binding affinity (0.5-2 nM)
    This comprehensive validation enabled identification of antibodies that specifically bound TF without inhibiting coagulation activity, making them ideal for imaging applications .

What mechanisms underlie the anticoagulant effects of TF monoclonal antibodies and how are these effects measured?

TF monoclonal antibodies exert anticoagulant effects through several mechanistic pathways that can be precisely measured using specialized assays:

Primary anticoagulant mechanisms:

  • Inhibition of TF-FVII(a) complex formation:

    • Antibodies target the FVII binding site on TF

    • Prevents the initial step in the extrinsic coagulation pathway

    • Results in complete inhibition of TF-initiated coagulation

  • Blockade of FX recruitment and activation:

    • Antibodies allow TF/FVIIa complex formation but interfere with FX binding

    • Specifically blocks the TF/FVIIa complex's ability to activate FX to FXa

    • Interrupts the coagulation cascade at a critical amplification step

  • Allosteric modulation of TF function:

    • Antibodies binding to non-active site regions induce conformational changes

    • Reduces catalytic efficiency of the TF/FVIIa complex

    • May selectively modify specific aspects of TF function

Quantitative assessment methods:

AssayMethodologyParameters MeasuredRelevance
Dilute Prothrombin Time (dPT)Measures clotting time in diluted plasma with exogenous TFProlongation of clotting timeIndicates inhibition of TF-initiated coagulation
FX Activation AssayMeasures conversion of FX to FXa using chromogenic substratesReduced rate of FXa generationDirectly assesses inhibition of FX activation by TF/FVIIa
Amidolytic Activity AssayMeasures catalytic activity of TF/FVIIa complexDecreased substrate cleavageEvaluates functional impact on enzyme complex activity
ThromboelastographyMeasures physical properties of clot formationAltered clot formation kineticsProvides global assessment of impact on coagulation
In vivo Thrombosis ModelsArterial/venous thrombosis in animal modelsThrombus size, prevention, lysis timeEvaluates physiological relevance of anticoagulant effects

Research example with clinical implications:
In a rabbit model of carotid artery thrombosis, the administration of AP-1 (an anti-TF monoclonal antibody) along with tissue plasminogen activator (TPA) significantly shortened lysis time from 44±8 minutes in control rabbits to 26±7 minutes in treated rabbits (P<0.01). Importantly, while reocclusion occurred in all control rabbits within 10±3 minutes, it occurred in only two of eight AP-1-treated rabbits and was significantly delayed (55-72 minutes). This demonstrates that TF exposure and activation of the extrinsic coagulation pathway play crucial roles in prolonging lysis time and mediating reocclusion after thrombolysis, suggesting that anti-TF monoclonal antibodies might be suitable as adjunctive therapy to TPA .

How do structural variations in TF monoclonal antibodies influence their functionality in different experimental and therapeutic contexts?

The structural characteristics of TF monoclonal antibodies profoundly impact their functionality across diverse applications, with rational design enabling optimization for specific research or therapeutic purposes:

Antibody format considerations:

FormatStructureSize (kDa)Functional ImplicationsOptimal Applications
Full IgGComplete antibody with Fc region150Long half-life, effector functions, slower tissue penetrationTherapeutic applications requiring extended duration, ADCC/CDC effects
F(ab')₂Bivalent fragment lacking Fc100Intermediate half-life, no effector functions, improved tissue penetrationImaging with extended circulation, therapeutic applications requiring Fc absence
FabMonovalent antigen-binding fragment50Short half-life, rapid tissue distribution, no avidity effectsRapid imaging, applications requiring quick clearance
scFvSingle-chain variable fragment25-30Very short half-life, excellent tissue penetration, potential instabilityMolecular imaging, building block for complex antibody formats
NanobodySingle-domain antibody fragment12-15Extremely rapid tissue penetration, high stabilityUltra-rapid imaging, accessing cryptic epitopes

Isotype and Fc engineering effects:

Different antibody isotypes (IgG1, IgG2, IgG3, IgG4) exhibit varying effector functions and half-lives. For therapeutic TF antibodies, isotype selection critically influences:

  • Complement activation potential

  • Fc receptor binding profiles

  • Antibody stability and aggregation propensity

  • Tissue distribution patterns

Advanced Fc engineering can further modulate these properties:

  • Enhanced ADCC through afucosylation or specific amino acid substitutions

  • Prolonged half-life via mutations enhancing FcRn binding

  • Reduced immunogenicity through deimmunization strategies

Case study in structural optimization:
The development of anti-TF antibody clone 1849 for glioma imaging illustrates structure-function considerations. For effective tumor visualization, researchers:

  • Selected antibodies with high specificity for human TF

  • Evaluated IgG format for appropriate circulation time allowing tumor accumulation

  • Optimized radiolabeling methods using 111In to maintain binding properties

  • Demonstrated efficient accumulation in TF-overexpressing intracranial tumors using both fluorescence and SPECT/CT imaging

Structure-based design implications:
Understanding the structural interactions between TF monoclonal antibodies and their target enables rational design of antibodies with tailored properties:

  • Antibodies targeting the FX binding site (like TF4A12) allow TF/FVIIa complex formation while blocking FX activation, providing selective anticoagulant effects

  • Non-inhibitory antibodies binding to epitopes outside functional domains can be utilized for imaging without disrupting coagulation

  • Next-generation formats like bispecific antibodies can simultaneously target TF and immune effector cells for enhanced therapeutic efficacy

These structural considerations allow researchers to precisely engineer TF antibodies for specific experimental or therapeutic applications, optimizing parameters such as tissue penetration, functional effects, and pharmacokinetic profile.

What are the challenges in developing non-inhibitory TF monoclonal antibodies for molecular imaging and how can these be overcome?

Developing non-inhibitory TF monoclonal antibodies for molecular imaging presents several challenges that require sophisticated solutions:

Fundamental challenges:

  • Limited non-functional epitope availability:

    • TF has a relatively small extracellular domain (~219 amino acids)

    • Many accessible epitopes overlap with coagulation function

    • The TF/FVIIa interaction surface occupies a substantial portion of the TF molecule

  • Binding affinity vs. functional neutralization trade-off:

    • High-affinity antibodies often disrupt TF function through allosteric effects

    • Lower-affinity antibodies may be insufficient for effective imaging

    • Conformational epitopes may be altered in different microenvironments

  • In vivo complexity factors:

    • Circulating soluble TF may compete for antibody binding

    • Encrypted (inactive) vs. decrypted (active) TF conformations

    • Species differences between preclinical models and humans

Strategic solutions and methodological approaches:

ChallengeAdvanced SolutionMethodologyValidation Approach
Identifying non-inhibitory epitopesStructural epitope mappingX-ray crystallography, cryo-EM, hydrogen-deuterium exchange MSCorrelation of epitope location with functional assays
Screening for non-inhibitory antibodiesHigh-throughput functional screeningParallel evaluation of binding and coagulation inhibitionScreening large antibody libraries against binding/function ratio
Optimizing binding without affecting functionAffinity maturation of non-inhibitory clonesPhage display with selective pressures, structure-guided mutationsSPR analysis (0.5-2nM affinity range) with functional testing
Reducing impact on hemostasisFormat engineering for rapid clearanceDevelopment of smaller fragments with shorter half-livesPharmacokinetic studies with coagulation monitoring
Species cross-reactivity issuesHumanized or human antibody developmentCDR grafting, transgenic animals, phage displayCross-species binding and functional studies

Successful case study:
Researchers developed a panel of murine monoclonal antibodies specific for human TF that did not inhibit TF-mediated blood coagulation using a modified RIMMS protocol. After primary ELISA screening, they verified binding to native human TF using flow cytometry with TF-expressing cells. Critical to success was their parallel evaluation approach:

  • Binding affinity determination using BIAcore (achieving 0.5-2 nM affinity)

  • Functional testing using blood coagulation assays to identify non-inhibitory antibodies

  • Further characterization of selected clones for stability and specificity

Current research directions:
The development of anti-TF 1849 monoclonal antibody illustrates progress in this field. This antibody efficiently accumulated in TF-overexpressing intracranial tumors when evaluated by both fluorescence and SPECT/CT imaging. The researchers proposed that immuno-SPECT with 111In-labeled anti-TF 1849 IgG could visualize biological characteristics of gliomas differently from existing imaging modalities, potentially helping evaluate malignancy grade and determine optimal biopsy locations .

This methodological approach demonstrates that with careful epitope selection, extensive screening, and advanced characterization techniques, it is possible to develop TF monoclonal antibodies suitable for molecular imaging applications.

What methodological approaches optimize TF monoclonal antibodies for cancer imaging, and what experimental data supports their efficacy?

Optimizing TF monoclonal antibodies for cancer imaging requires integrated methodological approaches spanning antibody engineering, conjugation chemistry, and imaging technologies:

Antibody optimization strategies:

  • Epitope selection:

    • Target TF epitopes overexpressed in cancer but not normal tissues

    • Select non-inhibitory epitopes to avoid systemic anticoagulation

    • Focus on tumor-specific TF conformations or post-translational modifications

  • Pharmacokinetic engineering:

    • Modify size through fragmentation (Fab, F(ab')₂, scFv) for optimal tumor penetration

    • Adjust clearance rate to match imaging modality requirements

    • Engineer for reduced binding to soluble TF to improve tumor-to-background ratio

  • Conjugation optimization:

    • Site-specific conjugation to prevent interference with antigen binding

    • Optimized chelator-to-antibody ratios for maximum signal

    • Selection of appropriate imaging moieties based on application needs

Experimental validation approaches:

Optimization PhaseExperimental MethodKey ParametersResearch Example
In vitro characterizationFlow cytometry, immunohistochemistryBinding specificity, cancer vs. normal tissue differentiationTF expression in gliomas increasing with WHO grade of malignancy
Affinity determinationSurface plasmon resonanceBinding kinetics, affinity constants (0.5-2 nM optimal range)High-affinity binding to human TF in BIAcore analyses
Ex vivo biodistributionTissue autoradiographyTumor uptake, organ distribution, tumor-to-background ratios-
In vivo imaging studiesSPECT/CT, PET/CT, optical imagingTumor visualization, quantitative uptake, pharmacokineticsEfficient accumulation of anti-TF 1849 IgG in TF-expressing intracranial tumors
Clinical translationFirst-in-human studiesSafety, dosimetry, diagnostic accuracy-

Supporting experimental evidence:

A comprehensive study using anti-human TF monoclonal antibody clone 1849 demonstrated several critical aspects of successful TF-targeted molecular imaging:

  • Correlation with tumor biology:
    Immunohistochemistry showed that TF expression in gliomas increased proportionally with WHO malignancy grade. TF was remarkably expressed in necrosis and pseudopalisading cells, histopathological hallmarks of glioblastoma multiforme (GBM) .

  • Multimodal imaging validation:
    Both fluorescence and SPECT/CT imaging studies confirmed that anti-TF 1849 IgG efficiently accumulated in TF-overexpressing intracranial tumors in mouse models .

  • Translational potential:
    The researchers proposed that immuno-SPECT with 111In-labeled anti-TF 1849 IgG could provide unique biological information different from existing imaging modalities, potentially helping to evaluate malignancy grade and determine biopsy locations in glioma patients .

Methodological advantages over conventional imaging:
TF-targeted monoclonal antibody imaging offers several advantages over traditional techniques:

  • Direct visualization of molecular pathology rather than anatomical features

  • Potential for earlier detection based on TF overexpression preceding morphological changes

  • Ability to distinguish tumor grades based on differential TF expression

  • Guidance for surgical intervention and biopsy targeting

The experimental data supports that properly optimized TF monoclonal antibodies can effectively visualize TF-expressing tumors, providing valuable biological information that complements existing imaging modalities.

How do TF monoclonal antibodies interact with the tumor microenvironment and what impact does this have on their therapeutic efficacy?

The interactions between TF monoclonal antibodies and the tumor microenvironment (TME) involve complex bidirectional relationships that significantly impact therapeutic outcomes:

TF distribution and functionality in the TME:

TF expression in tumors exhibits heterogeneous patterns across:

  • Tumor cells (variable expression levels correlating with malignancy grade)

  • Tumor vasculature (particularly in newly formed vessels)

  • Inflammatory cells within the tumor stroma

  • Tumor-associated microparticles and exosomes

This distribution creates multiple potential targets but also presents challenges for therapeutic coverage.

Key interaction mechanisms:

  • Blockade of TF-dependent coagulation in TME:

    • Inhibition of fibrin deposition that supports tumor growth

    • Reduction of platelet activation and aggregation

    • Prevention of thrombin generation that can activate protease-activated receptors (PARs)

  • Disruption of TF-mediated signaling:

    • Inhibition of TF/FVIIa-dependent signaling through PAR2

    • Reduction in pro-angiogenic factors production (VEGF, IL-8)

    • Suppression of tumor cell migration, invasion, and survival signals

  • Immune modulation within TME:

    • Fc-mediated recruitment of immune effector cells

    • Potential enhancement of tumor antigen presentation

    • Modification of the immunosuppressive microenvironment

  • Vascular effects:

    • Alterations in tumor vessel permeability

    • Changes in interstitial fluid pressure

    • Modified drug delivery to tumor tissue

TME factors limiting efficacy:

Limiting FactorMechanismPotential Solution
Poor antibody penetrationHigh interstitial pressure, abnormal vasculatureAntibody fragments, enhanced permeability strategies
Heterogeneous TF expressionSpatial and temporal variations in TF levelsCombination targeting, adaptive dosing regimens
Hypoxic regionsLimited antibody delivery to hypoxic areasHypoxia-activated antibody designs, combination approaches
Protease-rich environmentAntibody degradationProtease-resistant antibody engineering
Immunosuppressive factorsInhibition of effector functionsCombination with immune checkpoint inhibitors

Advanced therapeutic strategies exploiting TME interactions:

  • Antibody-drug conjugates (ADCs):
    TF-targeted ADCs deliver cytotoxic payloads directly to TF-expressing cells within the TME. The payload release mechanisms can be designed to exploit TME-specific conditions (reduced pH, elevated proteases) .

  • Bispecific antibodies:
    These can simultaneously target TF and immune effector cells, bringing cytotoxic immune cells in proximity to TF-expressing tumor cells and potentially overcoming immunosuppressive barriers .

  • Fc-engineered variants:
    Next-generation antibodies with enhanced Fc-mediated effector functions can more effectively engage immune cells within the TME, though their efficacy may vary depending on the specific tumor microenvironment .

  • Combination approaches:
    Combining TF monoclonal antibodies with agents that modify the TME (e.g., anti-angiogenic therapies, matrix-modifying agents, immune checkpoint inhibitors) may enhance therapeutic efficacy by addressing multiple hallmarks of cancer simultaneously.

The deep understanding of these complex interactions is driving the development of increasingly sophisticated TF-targeted therapeutic strategies that can navigate the challenges presented by the heterogeneous and dynamic tumor microenvironment.

What analytical methods best characterize the epitope specificity of TF monoclonal antibodies and how does this inform their research applications?

Comprehensive epitope characterization of TF monoclonal antibodies requires integrated analytical approaches that inform their optimal research applications:

Advanced epitope mapping technologies:

Analytical MethodTechnical ApproachResolution LevelSpecific Advantages
X-ray crystallographyCo-crystallization of Fab:TF complexesAtomic (1-3Å)Precise structural details of binding interface
Hydrogen-deuterium exchange mass spectrometry (HDX-MS)Analysis of deuterium incorporation patternsPeptide segments (5-20 amino acids)No crystallization required, solution-phase analysis
Cryo-electron microscopyVisualization of antibody:TF complexesMedium to high (3-5Å)Analysis of larger complexes, fewer sample constraints
Peptide array scanningBinding to overlapping peptide fragmentsLinear epitopesHigh-throughput, identifies continuous epitopes
Alanine scanning mutagenesisSystematic alanine substitution in TFSingle amino acidIdentifies critical binding residues
Cross-competition assaysCompetitive binding between antibodiesEpitope groupsClassified antibodies into minimum of five distinct groups
In silico epitope predictionComputational analysis of antibody:TF interfacesVariesRapid screening, guides experimental approaches

Correlation of epitope specificity with functional properties:

Detailed epitope mapping reveals critical structure-function relationships in TF monoclonal antibodies:

  • Factor VII binding site epitopes:

    • Antibodies targeting this region completely inhibit TF/FVIIa complex formation

    • Result in potent anticoagulant activity

    • Twenty-three hybridoma antibodies inhibited TF activity by blocking formation of the TF/FVII complex

  • Factor X interaction site epitopes:

    • Antibodies like TF4A12 block FX binding to TF/FVIIa complex without affecting FVII binding

    • Provide selective anticoagulant function

    • Mechanistically demonstrated through FX activation and amidolytic activity assays

  • Non-functional domain epitopes:

    • Antibodies binding outside coagulation-functional regions

    • Maintain binding without anticoagulant effects

    • Ideal for imaging applications requiring high target specificity without hemostatic disturbance

  • Conformational epitopes:

    • Recognize three-dimensional structures that may be cell-type specific

    • Potentially distinguish between encrypted vs. decrypted TF forms

    • Often have complex functional effects dependent on TF conformational state

Epitope mapping case study with methodological insights:
Researchers developed a library of twenty-four murine hybridomas secreting antibodies to human TF. Through extensive characterization using cross-competition assays, functional studies, and binding analyses, they categorized these antibodies into a minimum of five distinct groups based on their epitope recognition patterns. This systematic approach enabled them to correlate epitope specificity with functional properties, finding that twenty-three antibodies strongly inhibited TF activity by blocking TF/FVII complex formation .

Application-driven epitope selection:

Research ApplicationOptimal Epitope CharacteristicsAnalytical Validation Approach
Molecular imagingNon-inhibitory, tumor-enriched, high accessibilityImmunohistochemistry correlation with malignancy, in vivo imaging validation
Anticoagulant therapyFX binding site, predictable dose-responseCoagulation assays, animal models of thrombosis
Cancer research toolsHighly specific, detects various TF formsMulti-platform validation (WB, IP, IF, IHC, ELISA)
Antibody-drug conjugatesInternalizing epitopes, stable in circulationInternalization assays, stability studies

This methodologically rigorous approach to epitope characterization enables researchers to select or engineer TF monoclonal antibodies with precisely tailored properties for specific applications, maximizing their utility as research tools and therapeutic agents.

What factors determine the immunogenicity of therapeutic TF monoclonal antibodies and what experimental systems best predict clinical immunogenic responses?

Immunogenicity of therapeutic TF monoclonal antibodies is determined by multiple interacting factors that can be systematically evaluated using advanced predictive systems:

Critical determinants of immunogenicity:

  • Antibody structural factors:

    • Species origin (murine, chimeric, humanized, fully human)

    • Presence of T-cell epitopes within variable regions

    • Framework sequences that may contain immunogenic determinants

    • Post-translational modifications (non-human glycosylation patterns)

    • Aggregation propensity and conformational stability

  • Manufacturing-related factors:

    • Presence of host cell proteins or other process-related impurities

    • Protein modifications during production (oxidation, deamidation)

    • Formulation components affecting stability and aggregation

    • Storage conditions and handling procedures

  • Patient-related factors:

    • Genetic background and HLA haplotype

    • Prior exposure to similar biologics or environmental antigens

    • Disease state and concomitant immunomodulatory treatments

    • Route, dose, and frequency of administration

Predictive methodologies and their correlative value:

Prediction MethodExperimental ApproachPredictive ValueLimitations
In silico T-cell epitope predictionComputational analysis of binding to MHC-II moleculesModerateFails to consider protease cleavage sites, T cell precursor frequency, and competition factors
HLA binding assaysDirect measurement of peptide binding to MHC-II moleculesModerate-highBinding doesn't guarantee T cell recognition
Dendritic cell assaysAssessment of antigen processing and presentationGoodComplex to standardize, requires primary cells
T cell proliferation assaysMeasurement of T cell responses to antibody-derived peptidesHighResource-intensive, requires appropriate donor selection
PBMC cytokine release assaysEvaluation of cytokine secretion following stimulationHighDemonstrated with infliximab and rituximab variable regions
Transgenic mouse modelsHLA-transgenic mice expressing human MHC moleculesModerateSpecies differences in immune repertoire
Ex vivo immunogenicity assessmentPatient samples tested before clinical administrationHighLimited availability of appropriate samples

Integrated predictive approaches:
Most effective immunogenicity prediction employs multiple complementary methods:

  • Initial screening: In silico prediction algorithms to identify potential T-cell epitopes in TF antibody sequences

  • Intermediate validation: HLA binding assays to confirm computational predictions

  • Advanced evaluation: T cell proliferation and cytokine release assays using samples from diverse donors

    • Example: T cell epitopes in variable regions of therapeutic antibodies were shown to stimulate PBMCs to secrete various cytokines

    • The immunogenicity of secukinumab was assessed by examining T-cell proliferation

  • Pre-clinical integration: Transgenic animal models expressing human MHC molecules

Strategies to mitigate immunogenicity:

Risk FactorMitigation StrategyExperimental Validation
T-cell epitopesDeimmunization through targeted mutationsT cell assays before and after modification
Non-human sequencesHumanization or human antibody generationSequence homology analysis, immunogenicity assays
AggregationFormulation optimization, stability engineeringForced degradation studies, particle analysis
Manufacturing impuritiesProcess optimization, enhanced purificationHost cell protein analysis, impurity profiling

Case study in comprehensive immunogenicity assessment:
For therapeutic monoclonal antibodies including those targeting TF, researchers employ a multi-tiered approach:

  • Initial computational screening to identify potential T-cell epitopes

  • Experimental confirmation using HLA binding assays

  • Functional validation through T cell and PBMC stimulation assays

  • Ex vivo testing with patient samples when available

This methodologically rigorous approach provides a more complete prediction of potential immunogenicity than any single method alone, enabling the development of therapeutic TF monoclonal antibodies with reduced immunogenic potential .

How are advanced antibody engineering technologies being applied to TF monoclonal antibodies to enhance their research and therapeutic utility?

Advanced antibody engineering technologies are transforming TF monoclonal antibodies into increasingly sophisticated research and therapeutic tools:

Next-generation format engineering:

Engineered FormatStructural CharacteristicsTechnological AdvantagesApplications for TF Targeting
Bispecific antibodiesDual targeting of TF and a second antigenSimultaneous engagement of TF and immune cells or other targetsEnhanced tumor cell killing by recruiting immune effectors
Antibody fragments (Fab, scFv)Smaller size, lack of Fc regionImproved tissue penetration, rapid clearanceEnhanced tumor imaging, reduced immunogenicity
NanobodiesSingle-domain antibodies (~15 kDa)Exceptional stability, access to cryptic epitopesUltra-high resolution imaging, targeting sterically hindered TF epitopes
Fc-engineered variantsModified constant regionsEnhanced or silenced effector functionsOptimized ADCC/CDC for therapeutic applications, reduced effector function for imaging
pH-dependent binding antibodiesDifferential binding at varying pHHigher binding at tumor pH, cargo release in endosomesImproved tumor-specific targeting, reduced off-target effects

Payload conjugation innovations:

Antibody-drug conjugates (ADCs) represent a significant advancement for TF-targeted therapies:

  • Site-specific conjugation methods preserve antibody function

  • Novel linker chemistries enable controlled payload release

  • Optimization of drug-to-antibody ratios enhances therapeutic index

  • Dual-payload ADCs can deliver synergistic therapeutic agents

Affinity and specificity engineering:

  • Affinity maturation techniques:

    • Phage display with stringent selection conditions

    • Computational design of higher affinity variants

    • Structure-guided mutagenesis based on antibody-TF co-crystal structures

    • Directed evolution approaches using yeast or mammalian display systems

  • Specificity enhancement:

    • Engineering for preferential binding to tumor-associated TF forms

    • Developing conditional activation mechanisms responsive to tumor microenvironment

    • Creating antibodies sensitive to TF conformational states

Expression system innovations:

Expression SystemTechnological AdvantagesApplications
CHO and HEK cell linesHuman-like glycosylation, high production capacityTherapeutic antibodies requiring effector functions
Bacterial and yeast systemsSimple, high-performing, cost-effectiveResearch antibodies, fragments not requiring glycosylation
Transgenic animalsFully human antibodies, complex glycosylationHuman therapeutic antibodies with reduced immunogenicity
Cell-free expression systemsRapid production, site-specific incorporation of non-natural amino acidsSpecialized research antibodies, uniquely modified variants

Advanced engineering case studies:

  • Molecular imaging optimization:
    Anti-TF 1849 IgG demonstrated efficient accumulation in TF-overexpressing tumors during both fluorescence and SPECT/CT imaging studies. Further engineering could enhance this targeting through:

    • Development of smaller fragments for improved tumor penetration

    • Site-specific radioisotope conjugation for optimal imaging properties

    • Modifications to reduce binding to circulating TF

  • Therapeutic enhancement:
    Next-generation monoclonal antibodies are capitalizing on various mechanisms of action through improvements to enhance Fc-activity, with recognition that these mechanisms may vary in different tumor microenvironments. Antibody-drug conjugates (ADCs) have emerged as an important means to activate different mechanisms of action for improved therapeutic efficacy .

These advanced engineering approaches represent the cutting edge of TF monoclonal antibody development, enabling unprecedented precision in targeting TF for both research and therapeutic applications.

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.