ENG Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Monoclonal Antibody Fundamentals

Monoclonal antibodies are homogeneous proteins derived from a single B-cell clone, enabling targeted binding to specific epitopes . Their structure includes two Fab (antigen-binding) regions and an Fc (effector function) region, which can be engineered for enhanced efficacy or reduced immunogenicity .

FeatureDescription
SpecificityBind to a single epitope, reducing off-target effects .
Therapeutic MechanismsNeutralize pathogens, modulate immune responses, or deliver cytotoxic agents .
EngineeringAffinity maturation, bispecific/multi-specific designs, and Fc modifications .

Hypothetical ENG Monoclonal Antibody Applications

ApplicationPotential Use Case
Cancer TherapyTargeting angiogenic factors (e.g., Endoglin) to inhibit tumor blood vessel formation.
Diagnostic ToolsDetecting Endoglin expression in tissues to guide personalized therapies .
Autoimmune DiseasesModulating immune cell interactions via Endoglin pathways .

Monoclonal Antibody Engineering Advances

Recent innovations in mAb engineering include:

  • Bispecific Antibodies: Simultaneously targeting two epitopes (e.g., faricimab for VEGF-A and Ang-2) .

  • Antibody-Drug Conjugates (ADCs): Combining cytotoxic agents with antibodies (e.g., gemtuzumab ozogamicin for AML) .

  • Fc Modifications: Enhancing half-life (e.g., S228P mutation) or reducing immunogenicity (e.g., L234A/L235A mutations) .

Key Challenges in Antibody Development

ChallengeSolution
ImmunogenicityHumanizing antibodies via transgenic mice or phage display .
Aggregation/InstabilityEngineering hydrophobic residues (e.g., W30, F31, L56 mutations in MEDI1912) .
Tissue BindingOptimizing biophysical properties to reduce non-specific interactions .

Clinical and Regulatory Landscape

Over 200 monoclonal antibodies are approved or in development, addressing cancers, autoimmune diseases, and infections . For example:

AntibodyTargetIndicationApproved Year
TrastuzumabHER2Breast cancer1998
PembrolizumabPD-1Melanoma, NSCLC2014
FaricimabVEGF-A, Ang-2Diabetic macular edema2021
EvinacumabANGPTL3Hypercholesterolemia2021

Product Specs

Buffer
Liquid in PBS containing 50% glycerol, 0.5% BSA and 0.02% sodium azide.
Description

The ENG monoclonal antibody is produced by employing a synthesized peptide derived from human CD105 protein as an immunogen. B cells are isolated from the immunized mouse and then fused with myeloma cells. This results in hybridoma cells that are subsequently screened to identify the cell line producing the ENG antibody. The CD105 monoclonal antibody is affinity-purified from mouse ascites through affinity-chromatography using a specific immunogen. The purified antibody is an unconjugated IgG2b, Kappa. It demonstrates the ability to recognize human ENG protein in ELISA and IHC applications.

Endoglin (ENG), also known as CD105, is primarily expressed on endothelial cells and plays a critical role in angiogenesis. Endoglin functions as a co-receptor for TGF-β1 and TGF-β3, binding to these cytokines and enhancing their signaling activity in endothelial cells. It also interacts with other cell surface receptors, such as integrins, and extracellular matrix proteins, facilitating cell adhesion and migration. Furthermore, Endoglin has been shown to regulate vascular permeability, participate in vascular inflammation, and modulate cellular responses to oxidative stress.

Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 working days after receiving your orders. Delivery times may vary depending on the purchasing method or location. We encourage you to consult your local distributors for specific delivery details.
Synonyms
ENG; END; Endoglin; CD antigen CD105
Target Names
ENG
Uniprot No.

Target Background

Function
Endoglin, a glycoprotein present in the vascular endothelium, plays a pivotal role in regulating angiogenesis. It is essential for the normal structure and integrity of the adult vasculature, regulating the migration of vascular endothelial cells. Endoglin is crucial for normal extraembryonic angiogenesis and embryonic heart development. It may also regulate endothelial cell shape changes in response to blood flow, contributing to vascular remodeling and the establishment of normal vascular morphology during angiogenesis. Endoglin may play a significant role in the binding of endothelial cells to integrins and/or other RGD receptors. It functions as a TGF-beta coreceptor and participates in the TGF-beta/BMP signaling cascade, ultimately activating SMAD transcription factors. Endoglin is necessary for GDF2/BMP9 signaling through SMAD1 in endothelial cells and modulates TGFB1 signaling through SMAD3.
Gene References Into Functions
  1. Elevated soluble endoglin levels in cord blood are associated with the development of severe or moderate bronchopulmonary dysplasia in preterm infants with maternal preeclampsia. PMID: 30177044
  2. Mutations in ENG, ACVRL1, and SMAD4 genes lead to different phenotypes in hereditary hemorrhagic telangiectasia. PMID: 30251589
  3. High expression of CD105 is associated with Oral Invasive Carcinomas. PMID: 30049191
  4. The role of CD34 in determining the premalignant nature of oral submucous fibrosis (OSF) remains unclear, as all endothelial cells exhibited positivity for CD34. However, CD105 appears to be more specific, as it is linked to hypoxia-induced angiogenesis, which occurs in OSF due to hyalinization, suggesting that CD105 is a more specific marker for identifying neoangiogenesis in OSF. PMID: 30197335
  5. Data indicate that the extracellular domain of endoglin promotes specific platelet adhesion. PMID: 29080903
  6. ENG mutation carriers were more likely than ACVRL1 mutation carriers to have pulmonary arteriovenous malformations (pAVMs), multiple lesions, and undergo procedural intervention. The HHT severity score was significantly higher in ENG mutation carriers compared to ACVRL1 mutation carriers. PMID: 29048420
  7. Increased CD105 expression is associated with disease progression in phyllodes tumors. PMID: 29414396
  8. A heterozygous genetic variant, c.704dupC; p.Val236Glyfs*98, was identified in the ENG gene. The variant c.704dupC was not previously reported in the HHT Mutations Database. PMID: 29243366
  9. Endoglin levels are altered after stroke. PMID: 29287901
  10. A significant correlation was observed between CD105 expression and the survival rate of patients. A similar correlation was found between histological grades and TNM staging in CD105. High expression was associated with low survival; however, no significant correlation was observed between VEGF expression and survival, TNM staging, or histological grading. PMID: 29516931
  11. CD105 expression was associated with more aggressive tumor behavior, more advanced disease, and worse prognosis in clear cell renal cell carcinoma. PMID: 29286924
  12. Data suggest that in pancreatic cancer cells, the expression of ENG may be regulated by a pathway mediated by SMAD4. Additionally, ENG was found to be related to the spheroid-forming ability of cells and involved in the invasive capacity of pancreatic cancer cells. PMID: 29393426
  13. CD105 is expressed on endothelial cells of rhabdomyosarcoma and represents a valuable tool for quantifying neovascularization in this tumor. If confirmed by further studies, these results indicate that CD105 is a potential target for combined therapies in rhabdomyosarcoma. PMID: 29304781
  14. The increase in TGF-beta3 observed in inflammatory wound healing (WF) highlights its negative effect on wound healing. Conversely, the increased levels of soluble Endoglin in granulating WF affect leukocyte adhesion/transmigration through the endothelium, reducing the inflammatory response and promoting wound healing. PMID: 29065449
  15. Soluble endoglin levels could serve as a determinant of changes in walking abilities after a supervised treadmill training program in patients with peripheral artery disease. PMID: 28735679
  16. Serum levels are elevated in pre-eclampsia, not significantly affected by HIV status. PMID: 28627965
  17. High serum levels at 26-31 weeks of gestation were a risk factor for a small-for-gestational-age infant at 35-41 weeks. PMID: 28613009
  18. In primary hip OA, angiogenesis may be induced by a combined mechanism: hypoxia-related VEGF-dependent vasculogenesis and endothelial differentiation of the activated pluripotent cells released from the hyperplastic synovial cells layer. An endothelial mesenchymal transition is assumed to be involved in the fibrotic process. PMID: 27704157
  19. Findings demonstrate that Endoglin (CD105) expression not only demarcates a cancer stem cell subpopulation but also confers self-renewal ability and contributes to chemoresistance in renal cell carcinoma (RCC). PMID: 28793246
  20. In patients with hypertensive disorders of pregnancy, those in the highest tertile of mean arterial pressure had the highest serum levels of sFlt1 and sEng. PMID: 28609171
  21. BMP9 interacts with a hydrophobic surface of the N-terminal orphan domain of ENG, which adopts a new duplicated fold generated by circular permutation. PMID: 28564608
  22. Tale of Two Endoglins: How Does Tail-Less Soluble Endoglin Deregulate Lung Development? PMID: 28960105
  23. The variant lies in a novel binding-site for the transcription factor Sp1, known to be involved in the regulation of ENG and ACVRL1 transcription. PMID: 29305977
  24. Mobilization of the CD44/CD105 positive synovial cells appears to play a role in the genesis of hip osteoarthritis. PMID: 27803113
  25. Circulating tissue transglutaminase is associated with sFlt-1, soluble endoglin, and VEGF in the maternal circulation of preeclampsia patients, suggesting that tTG may have a role in the pathogenesis of PE. PMID: 27169826
  26. An altered antiangiogenic state due to altered circulating sEng leads to Preeclampsia. PMID: 27067718
  27. Gestation-adjusted sEng, sFlt-1, and PlGF levels were lower in women who later suffered miscarriage compared with unaffected pregnancies. PMID: 27664209
  28. This meta-analysis demonstrated that CD105 overexpression correlates with higher WHO grade and poor survival. PMID: 26884265
  29. Nuclear survivin expression correlates with endoglin-assessed microvascularisation in laryngeal squamous cell carcinoma. PMID: 28446541
  30. Dendritic cells loaded with lysates derived from CD105+ human renal cell carcinoma cancer stem cells (CSCs) induced more functionally specific active T cells and specific antibodies against CSCs, and clearly depressed tumor growth in mice. PMID: 28621442
  31. No relationship was reported between serum endoglin levels and ovarian cancer microvessel density and tumor endoglin expression. PMID: 27312585
  32. TGF-beta type I, II, and III receptors were all identified in pregnant serum; all were substantially elevated in early-onset but not late-onset PE. Endoglin was increased in both subtypes. PMID: 28633389
  33. A series of pulmonary arterial hypertension patients with mutations in the ENG gene, some not previously described, exhibited clinical and hemodynamic alterations. These findings suggest that the presence of these mutations may be associated with the severity of the disease. PMID: 27260700
  34. CD105(+) blasts are endowed with superior leukemogenic activity compared to the CD105(-) population. PMID: 28351936
  35. Several germline variants in Hamartomatous Polyposis Syndrome genes were detected, including three in ENG, two in BMPR1A, one in PTEN, and one in SMAD4. While some of the detected variants have been reported previously, none could be definitively classified as pathogenic or likely pathogenic. PMID: 27146957
  36. A 9q33.3q34.11 microdeletion encompassing the ENG gene was identified in four patients with intellectual disability, epilepsy, nail dysplasia, and bone malformations. PMID: 26395556
  37. Endoglin plays an important role in VSMC recruitment and blood vessel maturation during angiogenesis. PMID: 28450296
  38. Plasma sEng levels were low in patients with coronary artery disease, particularly 3-vessel disease, and were inversely associated with the severity of coronary atherosclerosis. PMID: 27789477
  39. Soluble endoglin did not vary over the pregnancy course or between gestational hypertension, preeclampsia, and control groups. PMID: 27793555
  40. Serum and placental LXR-alpha and endoglin levels were significantly higher in patients with preeclampsia compared to the control group. PMID: 27736929
  41. High concentrations of sEng in plasma, in combination with a high-fat diet, induce the simultaneous activation of proinflammatory, pro-oxidative, and vasoprotective mechanisms in the mouse aorta. The balance of these biological processes determines whether the final endothelial phenotype is adaptive or maladaptive. PMID: 27721318
  42. Findings suggest a stronger chondrogenic potential of CD105(+) SMSCs compared to CD105(-) SMSCs. CD105 enhances chondrogenesis of SMSCs by regulating the TGF-beta/Smad2 signaling pathway but not Smad1/5. This study provides a better understanding of CD105's role in chondrogenic differentiation. PMID: 27107692
  43. The novel ENG c.-58G/A substitution in the Endoglin promoter co-segregates with Hereditary hemorrhagic telangiectasia symptoms in a family and appears to affect the transcriptional regulation of the gene, resulting in reduced Endoglin expression. PMID: 28231770
  44. sEng treatment resulted in an activation of NF-kappaB, IL-6, suggesting activation of a pro-inflammatory phenotype in endothelial cells. PMID: 28336397
  45. In adolescents with type 1 diabetes mellitus (T1DM), soluble endoglin concentrations might increase in parallel with the deterioration in endothelial function before subclinical structural vascular alterations. PMID: 27097763
  46. Akt levels were reduced in preeclamptic placentas relative to preterm controls. Inhibition of PI3K/Akt resulted in significantly elevated soluble endoglin release from endothelial cells, had no effect on MMP14 mRNA expression but resulted in significantly reduced TIMP3. In contrast, inhibiting PI3K/Akt in placental explants or primary trophoblast did not alter soluble endoglin release. PMID: 27155335
  47. Serum levels are associated with the severity of gestational hypertension and pre-eclampsia. PMID: 28121958
  48. The TGFbeta1 coreceptor Eng selectively regulates the expression of multiple transient receptor potential channels in the setting of left or right ventricular pressure overload. PMID: 27614169
  49. Increased ENG gene expression is associated with the risk of hepatocellular carcinoma. PMID: 27268609
  50. Higher levels of circulating CD105 are related to adverse pathological features among patients with oral squamous cell carcinoma. PMID: 26334621

Show More

Hide All

Database Links

HGNC: 3349

OMIM: 131195

KEGG: hsa:2022

STRING: 9606.ENSP00000362299

UniGene: Hs.76753

Involvement In Disease
Telangiectasia, hereditary hemorrhagic, 1 (HHT1)
Subcellular Location
Cell membrane; Single-pass type I membrane protein.
Tissue Specificity
Detected on umbilical veil endothelial cells. Detected in placenta (at protein level). Detected on endothelial cells.

Q&A

What distinguishes monoclonal antibodies from polyclonal antibodies in research applications?

Monoclonal antibodies (mAbs) are homogeneous, mono-specific molecules produced from a single B-cell clone, whereas polyclonal antibodies (pAbs) are heterogeneous mixtures derived from multiple B-cell clones. This fundamental difference results in distinct research applications:

Monoclonal antibodies:

  • Arise from single cell clones, providing consistent specificity to a single epitope

  • Offer high reproducibility across experiments and batches

  • Enable precise targeting of specific protein conformations or modifications

  • Typically require longer development time (several months to years)

  • Produced through hybridoma technology or recombinant DNA methods

Polyclonal antibodies:

  • Generated from multiple B-cell clones, recognizing various epitopes on the same antigen

  • Can be produced more rapidly (several months) and at lower cost

  • Maintain stability across broader pH and salt concentration ranges

  • Recognize multiple linear epitopes with minimal conformational changes

  • Typically form larger precipitating lattices in immunoprecipitation applications

The choice between mAbs and pAbs depends on experimental requirements for specificity, sensitivity, and application context. For precisely targeted molecular interactions or therapeutic applications, mAbs provide superior consistency and specificity, while pAbs offer advantages in applications requiring robust antigen recognition across multiple epitopes .

How do monoclonal antibodies function as immune system modulators?

Monoclonal antibodies function as immune system modulators through multiple mechanisms that can be strategically engineered for specific research or therapeutic applications:

  • Antigen binding and flagging: mAbs precisely attach to specific antigens on target cells (such as cancer cells), serving as flags that attract immune system components. This binding occurs through the variable regions of the antibody that are engineered for high specificity and affinity to particular molecular targets .

  • Complement activation: Upon binding to target antigens, certain mAbs activate the complement system, triggering a cascade of enzymatic reactions leading to the formation of the membrane attack complex that creates pores in the target cell membrane, resulting in cell lysis .

  • Antibody-dependent cellular cytotoxicity (ADCC): mAbs can engage Fc receptors on immune effector cells such as natural killer cells, macrophages, and neutrophils. This interaction activates these cells to attack and destroy the antibody-coated target cells through various cytotoxic mechanisms .

  • Blockade of molecular signaling: mAbs can be designed to inhibit receptor-ligand interactions essential for cell survival or proliferation. For example, some therapeutic mAbs target growth factors or their receptors to block cancer cell proliferation signals .

  • Immune checkpoint modulation: A specialized class of mAbs targets immune checkpoint molecules (such as CTLA-4, PD-1, or PD-L1) to enhance anti-tumor immune responses by removing inhibitory signals that limit T-cell activation .

Each of these mechanisms can be optimized through antibody engineering techniques, including Fc region modifications to enhance effector functions or alterations to binding domains to improve specificity and affinity .

How has the historical development of monoclonal antibody technology shaped current research approaches?

The historical trajectory of monoclonal antibody development has fundamentally shaped current research methodologies through several key innovations:

The foundation was established in 1975 when Köhler and Milstein first described hybridoma technology in Nature, later earning them a Nobel Prize. This breakthrough method fused antibody-producing spleen cells from immunized mice with immortal myeloma cells to create hybrid cell lines capable of continuous antibody production . This technology transformed antibody research by enabling the consistent production of homogeneous antibodies with defined specificities.

The evolution of mAb development can be traced through distinct phases:

  • First-generation mAbs (1980s): Purely murine antibodies that faced significant limitations in human applications due to immunogenicity and short half-lives. These mouse-derived antibodies frequently triggered human anti-mouse antibody (HAMA) responses, limiting their therapeutic potential .

  • Chimeric and humanized antibodies (1990s): Antibody engineering techniques created chimeric antibodies (human constant regions with murine variable regions) and humanized antibodies (only complementarity-determining regions from murine sources), reducing immunogenicity while maintaining specificity .

  • Fully human antibodies (2000s): Development of transgenic mice containing human antibody genes and phage display libraries enabled production of fully human antibodies, dramatically reducing immunogenicity issues .

  • Fragment-based and alternative scaffold approaches (2010s onward): Engineering of antibody fragments (Fab, scFv) and novel binding proteins expanded the toolbox for research and therapeutic applications .

This historical progression has influenced current research methodologies by emphasizing:

  • Recombinant DNA approaches over traditional immunization

  • In vitro selection strategies for specificity engineering

  • Rational design principles for optimizing binding and functional properties

  • Integration of computational modeling with experimental validation

The FDA approval trajectory reflects this evolution, with approximately 47 mAb products approved by 2015, and projections suggesting around 70 products would be on market by 2020, with global trade reaching approximately $125 billion .

What are the critical quality control parameters for monoclonal antibody production in research settings?

Rigorous quality control is essential for monoclonal antibody production in research settings to ensure reproducibility and reliability of experimental results. Key quality control parameters include:

  • Specificity assessment:

    • Cross-reactivity testing against related and unrelated antigens

    • Epitope mapping to confirm consistent target recognition

    • Competitive binding assays to verify binding site specificity

    • Western blot, ELISA, and immunohistochemistry validation across multiple sample types

  • Functional characterization:

    • Affinity determination through surface plasmon resonance or biolayer interferometry

    • Neutralization potency assessment for blocking antibodies

    • Effector function analysis (ADCC, CDC) for Fc-dependent activities

    • Thermal stability and pH sensitivity profiling

  • Physicochemical properties:

    • Size exclusion chromatography to assess aggregation levels

    • Charge variant analysis using ion exchange chromatography

    • Glycosylation profile analysis via mass spectrometry

    • Endotoxin testing to ensure preparations are pyrogen-free

  • Production consistency:

    • Clone stability monitoring across multiple passages

    • Batch-to-batch comparison of critical quality attributes

    • Growth kinetics and productivity assessment

    • Media component analysis to identify critical parameters

  • Purity verification:

    • SDS-PAGE and capillary electrophoresis for homogeneity assessment

    • Host cell protein quantification (typically <100 ppm)

    • Residual DNA quantification (<10 ng per dose)

    • Mycoplasma and viral contamination testing

Implementation of these quality control parameters should follow a risk-based approach, where critical attributes that impact the antibody's intended research application receive the most rigorous testing. Documentation of all quality parameters in standardized formats ensures reproducibility across laboratory settings and facilitates troubleshooting when experimental inconsistencies arise .

How do contemporary antibody engineering techniques overcome limitations of traditional hybridoma technology?

Contemporary antibody engineering techniques have revolutionized monoclonal antibody development by addressing several fundamental limitations of traditional hybridoma technology:

Limitation: Restricted species origin and immunogenicity

  • Engineering solution: Humanization techniques transform mouse-derived antibodies by grafting complementarity-determining regions (CDRs) onto human antibody frameworks, dramatically reducing immunogenicity while preserving antigen specificity .

  • Advanced approach: Transgenic mice with human immunoglobulin genes produce fully human antibodies through conventional immunization, eliminating humanization requirements entirely .

Limitation: Dependency on immune response

  • Engineering solution: Phage display technology bypasses immunization by presenting antibody fragments on bacteriophage surfaces, allowing in vitro selection against virtually any antigen, including toxic or self-antigens that cannot elicit natural immune responses .

  • Advanced approach: Cell-free display systems (ribosome, mRNA, and yeast display) offer alternative platforms for screening massive antibody libraries under controlled conditions .

Limitation: Full-size antibody constraints

  • Engineering solution: Recombinant fragment technologies produce Fab, scFv (single-chain variable fragments), and nanobodies with enhanced tissue penetration and simplified production requirements .

  • Advanced approach: Bi-specific antibodies and alternative scaffold proteins enable novel functionalities impossible with conventional antibody structures .

Limitation: Limited functional properties

  • Engineering solution: Fc engineering modifies effector functions through amino acid substitutions or glycoengineering, creating antibodies with enhanced or silenced ADCC/CDC activities as required .

  • Advanced approach: Directed evolution through iterative mutagenesis and selection cycles optimizes binding affinity, stability, and manufacturability beyond naturally occurring limits .

Limitation: Challenging manufacturing

  • Engineering solution: Mammalian expression systems (CHO, HEK293) with optimized vectors and selection markers increase expression yields by over ten-fold compared to hybridoma cultures .

  • Advanced approach: Cell-line engineering removes problematic glycosylation patterns and introduces beneficial modifications to enhance consistency and functional properties .

These engineering approaches have dramatically expanded the therapeutic potential of antibodies, enabling precise targeting of previously "undruggable" targets and creating molecules with novel mechanisms of action that were impossible with traditional hybridoma technology .

What methodological approaches effectively address the challenge of antibody immunogenicity in therapeutic applications?

Addressing antibody immunogenicity represents a critical challenge in therapeutic monoclonal antibody development. Researchers employ multiple methodological approaches to minimize immunogenic potential while maintaining therapeutic efficacy:

Methodological validation typically involves:

  • In silico T-cell epitope prediction

  • Ex vivo human T-cell assays using donor peripheral blood mononuclear cells

  • Non-human primate studies to assess comparative immunogenicity

  • Careful monitoring of anti-drug antibody development in clinical trials

The effectiveness of these approaches is demonstrated by the dramatic reduction in immunogenicity from first-generation murine antibodies (which showed >80% anti-drug antibody development in patients) to modern fully human antibodies (typically <1-10% immunogenicity rates) .

How can researchers optimize monoclonal antibody fragments for enhanced tissue penetration in solid tumor research?

Optimizing monoclonal antibody fragments for improved solid tumor penetration requires systematic engineering approaches addressing size, binding properties, and pharmacokinetics:

  • Fragment size optimization:

    • Standard IgG antibodies (~150 kDa) show limited tumor penetration due to their large size, which restricts diffusion through dense tumor tissue and abnormal vasculature

    • Smaller fragments demonstrate inverse correlation between size and tumor penetration:

      • Fab fragments (~50 kDa): 3-5× improved penetration versus full IgG

      • scFv (~25 kDa): 5-8× improved penetration

      • sdAb/nanobodies (~15 kDa): 8-10× improved penetration

    • Optimization involves balancing size reduction against stability and manufacturing considerations

  • Affinity modulation strategies:

    • Counter-intuitively, extremely high affinity can impair tumor penetration through a "binding site barrier" effect where antibodies bind strongly to the first antigen encountered

    • Methodological approach:

      • Engineer moderate affinity variants (KD ~10⁻⁸-10⁻⁷ M) through directed mutagenesis of CDR residues

      • Test penetration using 3D tumor spheroid models

      • Compare high and moderate affinity variants at equivalent doses

    • Implementation of pH-dependent binding can allow efficient tumor release and recycling

  • Surface charge engineering:

    • Positive surface charge increases non-specific tissue interactions

    • Systematic substitution of surface lysine and arginine residues with neutral or negatively charged amino acids significantly improves tumor-specific localization

    • Isoelectric point reduction from pI 8-9 to pI 5-7 can improve tumor:blood ratios by 2-4 fold

  • Half-life extension technologies:

    • While smaller fragments improve penetration, they typically exhibit rapid renal clearance

    • Implementation options:

      • Site-specific PEGylation at engineered cysteine residues

      • Fusion to albumin-binding domains or albumin itself

      • Fc-fusion constructs with engineered Fc domains

      • Implementation of the "DART" (Dual-Affinity Re-Targeting) format

    • Each approach requires empirical optimization for the specific target and tumor type

  • Multi-specific formatting:

    • Bispecific constructs can target both tumor antigens and components of tumor vasculature (e.g., VEGFR2)

    • Methodological format options include:

      • Tandem scFvs

      • Diabodies

      • TandAbs

      • DVD-Ig (dual-variable domain immunoglobulins)

    • This approach enhances localization while potentially addressing tumor heterogeneity

Experimental validation should employ physiologically relevant models including:

  • 3D tumor spheroids for initial penetration assessment

  • Patient-derived xenografts for translational relevance

  • Intravital microscopy for real-time visualization of penetration dynamics

These optimization strategies have collectively demonstrated the potential to improve tumor:blood ratios by 5-20 fold compared to conventional antibodies while maintaining target specificity .

What methodological approaches resolve conflicting data when assessing monoclonal antibody specificity?

Resolving conflicting specificity data for monoclonal antibodies requires systematic troubleshooting and orthogonal validation approaches. Researchers should implement the following methodological framework:

  • Comprehensive epitope characterization:

    • Map the precise epitope using hydrogen-deuterium exchange mass spectrometry or X-ray crystallography to determine if apparent cross-reactivity stems from conserved epitopes across related proteins

    • Implement alanine-scanning mutagenesis to identify critical binding residues

    • Compare epitope conservation across species when cross-reactivity appears in unexpected sample types

    • Create epitope-specific peptide arrays to confirm binding specificity against potential cross-reactive sequences

  • Multi-platform validation protocol:

    • Implement at least three orthogonal techniques to assess specificity:

      • Western blotting (denatured proteins)

      • ELISA/SPR (native proteins)

      • Immunohistochemistry/Immunofluorescence (tissue context)

      • Immunoprecipitation-mass spectrometry (definitive target identification)

    • Specifically test using knockout/knockdown controls alongside wild-type samples

    • Include competition assays with known ligands or alternate antibodies to the same target

  • Sample preparation variables assessment:

    • Systematically evaluate how different sample preparations affect epitope accessibility:

      • Fixation methods (formalin, methanol, acetone)

      • Antigen retrieval techniques (heat-induced, enzymatic, pH variations)

      • Blocking reagents (BSA, serum, commercial blockers)

      • Detergent types and concentrations for membrane proteins

    • Document all preparation conditions thoroughly to identify sources of variation

  • Clone and production batch verification:

    • Sequence verify the antibody-producing hybridoma to confirm no genetic drift

    • Test multiple production batches to identify potential manufacturing variables

    • Compare original hybridoma-derived antibody with recombinantly produced versions

    • Implement reference standards for comparative analysis across experiments

  • Advanced analytical resolution approaches:

    • Employ analytical SEC (size exclusion chromatography) to assess aggregation status

    • Implement charge variant analysis via ion-exchange chromatography

    • Utilize glycoform profiling to identify post-translational differences between batches

    • Consider mass spectrometric analysis of the antibody itself to confirm sequence and modifications

When conflicting data persist despite these approaches, researchers should consider developing new antibodies using alternative immunization strategies or in vitro display technologies, potentially targeting different epitopes on the same protein . The most definitive validation comes from parallel testing in systems with genetic knockouts of the target protein, which should completely eliminate specific binding .

How can researchers engineer monoclonal antibodies for optimal blood-brain barrier penetration in neurodegenerative disease research?

Engineering monoclonal antibodies for enhanced blood-brain barrier (BBB) penetration requires specialized strategies that address the unique challenges of targeting the central nervous system (CNS). Researchers can implement several methodological approaches to optimize antibody delivery across this restrictive barrier:

Experimental validation should employ a multi-faceted approach:

  • Quantitative biodistribution studies using radiolabeled antibodies

  • Fluorescence microscopy with co-localization to verify target engagement

  • CSF/plasma ratio determination as a surrogate for BBB penetration

  • PET imaging for real-time assessment of brain penetration in vivo

  • Functional assays demonstrating target engagement (e.g., plaque reduction)

These engineering strategies collectively can improve brain exposure by 10-100 fold compared to conventional antibodies, though absolute brain levels typically remain at 0.1-1% of plasma concentration even with optimized constructs .

How do display technologies enable the development of high-affinity monoclonal antibodies against challenging targets?

Display technologies have revolutionized the development of high-affinity monoclonal antibodies against challenging targets by enabling in vitro selection without dependency on animal immune responses. These platforms offer methodological advantages that address limitations of traditional hybridoma approaches:

  • Phage display technology:

    • Methodology: Antibody fragments (scFv or Fab) are genetically fused to bacteriophage coat proteins, creating a physical linkage between phenotype (binding) and genotype (sequence)

    • Library construction: Diverse libraries (10^9-10^11 members) are created through:

      • Synthetic diversity in complementarity-determining regions (CDRs)

      • Natural diversity from human B-cell repertoires

      • Semi-synthetic approaches combining framework stability with designed CDR diversity

    • Selection process: Iterative rounds of:

      • Binding to immobilized target

      • Washing to remove non-binders

      • Elution of specific binders

      • Amplification in bacterial hosts

    • Applications: Particularly effective for self-antigens, toxic targets, and conserved epitopes that fail to elicit immune responses in animals

  • Yeast display systems:

    • Advantages over phage:

      • Eukaryotic protein folding and post-translational modifications

      • Compatibility with fluorescence-activated cell sorting (FACS)

      • Quantitative screening based on binding affinity

    • Selection methodology:

      • Antibody fragments displayed on yeast cell surface as fusions to Aga2p protein

      • Dual-color FACS allows simultaneous selection for expression and binding

      • Progressive reduction in target concentration enables affinity maturation

    • Demonstrated capability: Generation of antibodies with sub-picomolar affinities through directed evolution

  • Ribosome and mRNA display:

    • Cell-free advantage: Libraries of 10^12-10^14 diversity, exceeding cellular transformation limits

    • Methodology:

      • Translation complexes link nascent antibody fragments to their encoding mRNA

      • Selection occurs entirely in vitro under controllable conditions

      • Reverse transcription and PCR amplification recover selected sequences

    • Applications: Particularly suited for affinity maturation through error-prone PCR and stringent selection

  • Mammalian display systems:

    • Unique benefits:

      • Full-length antibody display with authentic glycosylation

      • Direct assessment of manufacturability characteristics

      • Selection for both binding and expression properties

    • Methodology:

      • Antibodies displayed on mammalian cell surface with co-expressed reporter genes

      • FACS-based selection for binding and expression levels

      • Direct transition from selected clones to production cell lines

These display technologies enable methodological approaches impossible with hybridoma technology:

  • Epitope-focused selection strategies:

    • Sequential positive/negative selections against related antigens to identify rare epitope-specific binders

    • Competitive selections in the presence of known ligands or antibodies

    • Cross-species consensus epitope targeting for conserved functional domains

  • Affinity maturation protocols:

    • Creation of targeted mutagenesis libraries focusing on CDRs

    • Progressive reduction in target concentration (101-104 fold) across selection rounds

    • Off-rate selections using excess unlabeled competitor

    • Temperature challenge to select thermostable high-affinity variants

Through these methodologies, display technologies have successfully generated antibodies against traditionally challenging targets including:

  • G-protein coupled receptors in native conformations

  • Ion channels with limited extracellular domains

  • Transient protein-protein interaction interfaces

  • Conformational epitopes on intrinsically disordered proteins

What strategies effectively optimize the pharmacokinetic properties of engineered monoclonal antibodies?

Optimizing the pharmacokinetic (PK) properties of engineered monoclonal antibodies requires systematic modification of molecular characteristics that influence absorption, distribution, metabolism, and elimination. Researchers can implement the following methodological strategies:

  • Half-life extension engineering:

    • Fc engineering approaches:

      • Implement specific mutations in the CH2-CH3 interface (M252Y/S254T/T256E, "YTE" mutations) to enhance binding to the neonatal Fc receptor (FcRn) at endosomal pH (~6.0) while maintaining weak binding at physiological pH (~7.4)

      • These modifications can extend half-life 3-4 fold by enhancing antibody recycling through the FcRn salvage pathway

      • Histidine substitutions at positions 433/434 provide pH-dependent FcRn binding

    • Albumin-binding strategies:

      • Fusion of albumin-binding domains derived from streptococcal protein G

      • Direct fusion to albumin

      • These approaches leverage albumin's 19-day half-life in humans

  • Tissue distribution optimization:

    • Size manipulation:

      • Smaller fragments (Fab, scFv) exhibit enhanced tissue penetration but shorter half-lives

      • Larger formats (IgG, fusion proteins) show extended circulation but limited tissue access

      • Bispecific constructs can balance these properties through careful domain arrangement

    • Charge modification:

      • Systematic reduction of isoelectric point (pI) from 8-9 to 5-7 through surface residue substitutions

      • Removal of positively charged patches that contribute to non-specific tissue binding

      • Introduction of specific glycosylation patterns to modify charge distribution

  • Clearance route modulation:

    • Renal filtration management:

      • Molecules below ~60 kDa undergo significant renal filtration

      • PEGylation at specific sites can increase hydrodynamic radius without proportional mass increase

      • Multimerization strategies (diabodies, triabodies) increase effective size

    • Glycoengineering approaches:

      • Removal of terminal galactose or sialic acid residues to reduce asialoglycoprotein receptor-mediated clearance

      • Elimination of high-mannose glycans that accelerate clearance through mannose receptors

      • Site-specific glycosylation engineering through introduction or removal of N-glycosylation sites

  • Administration route optimization:

    • Subcutaneous delivery enhancement:

      • Co-formulation with hyaluronidase to facilitate absorption from subcutaneous space

      • Concentration optimization (typically 100-150 mg/mL) to balance viscosity and volume

      • Modification of interaction with extracellular matrix components through charge engineering

    • Site-specific delivery approaches:

      • Development of depot formulations using hydrogels or crystallization approaches

      • Implementation of targeting moieties for tissue-specific accumulation

      • pH or temperature-sensitive formulations that respond to microenvironmental conditions

  • Stability engineering for consistent PK:

    • Thermal stability enhancement:

      • Introduction of disulfide bonds at strategic positions

      • Framework stabilizing mutations identified through computational design

      • CDR grafting onto stable frameworks while maintaining specificity

    • Aggregation resistance:

      • Identification and modification of aggregation-prone regions

      • Surface hydrophobicity reduction through targeted mutations

      • Engineering to reduce chemical degradation pathways (deamidation, oxidation, isomerization)

Implementation of these strategies requires iterative optimization with attention to potential trade-offs between pharmacokinetic properties and other critical qualities including immunogenicity, manufacturing feasibility, and target binding .

How can multispecific antibody platforms be leveraged to address complex disease mechanisms in research?

Multispecific antibody platforms represent a revolutionary approach for addressing complex disease mechanisms by enabling simultaneous engagement of multiple targets within a single molecule. These engineered constructs overcome fundamental limitations of conventional monoclonal antibodies and offer unique research capabilities:

  • Bispecific T-cell engager (BiTE) platforms for cancer research:

    • Methodological design: These molecules simultaneously bind CD3 on T cells and tumor-associated antigens, creating a physical bridge that redirects T-cell cytotoxicity

    • Format optimization:

      • scFv-based constructs (e.g., blinatumomab) offer simplicity but short half-life

      • Fc-containing formats provide extended circulation while maintaining dual targeting

      • Domain orientation and linker optimization critically influence activity

    • Research applications:

      • Investigation of MHC-independent T-cell activation mechanisms

      • Comparative analysis of different tumor antigens for T-cell redirection

      • Study of resistance mechanisms to immune checkpoint inhibition

  • Dual targeting approaches for signaling pathway modulation:

    • Methodological rationale: Simultaneous blockade of multiple nodes in signaling networks overcomes redundancy and resistance mechanisms

    • Format options:

      • IgG-scFv fusions targeting two different receptors

      • DVD-Ig (dual-variable domain immunoglobulins) with tandem variable domains

      • CrossMAb technology for asymmetric heavy chain pairing

    • Research applications:

      • Dissection of compensatory signaling networks

      • Identification of synergistic receptor combinations

      • Development of models with reduced resistance to targeted therapies

  • Multispecific approaches for targeting protein complexes:

    • Methodological advantage: These constructs can selectively target protein-protein interactions or specific complex conformations

    • Implementation strategies:

      • Avidity-driven binding requiring engagement of both epitopes

      • Conditional activation dependent on dual binding

      • Targeting of neo-epitopes at protein interfaces

    • Research applications:

      • Identification and validation of protein complex-specific biology

      • Differentiation between monomeric and assembled states

      • Development of tools to study transient molecular interactions

  • Blood-brain barrier (BBB) crossing strategies:

    • Methodological approach: One binding arm targets BBB transporters (e.g., transferrin receptor) while the other engages the CNS disease target

    • Format considerations:

      • Binding affinity to transport receptor requires careful optimization

      • Brain-to-blood ratio depends on relative affinities to both targets

      • Molecular size influences transcytosis efficiency

    • Research applications:

      • In vivo target engagement studies in CNS disease models

      • Comparative assessment of different BBB transport mechanisms

      • Development of tools for studying CNS biology

  • Trispecific and higher-order multispecifics:

    • Advanced architectural platforms:

      • TRIOMAB technology (three functional binding sites)

      • IgG-scFv2 fusions with dual C-terminal scFvs

      • Knobs-into-holes technology enabling asymmetric designs

    • Research applications:

      • Simultaneous blockade of multiple escape pathways

      • Engineering of artificial cellular interactions involving multiple cell types

      • Development of synthetic cellular communication systems

Implementation considerations for research applications include:

  • Expression system selection (mammalian vs. microbial)

  • Purification strategy development for homogeneous preparations

  • Structural and biophysical characterization

  • Functional validation in physiologically relevant systems

These multispecific platforms enable unprecedented experimental approaches for studying complex biological systems by creating novel molecular interactions impossible with conventional monoclonal antibodies, facilitating research into emergent properties of biological networks .

What methodological approaches can resolve inconsistent monoclonal antibody performance across different experimental platforms?

Inconsistent monoclonal antibody performance across experimental platforms is a common challenge that requires systematic troubleshooting. Researchers can implement the following methodological framework to identify and resolve discrepancies:

  • Epitope accessibility assessment:

    • Methodological approach: Different platforms expose antigens in varying conformational states

      • Western blotting: predominantly linear epitopes under denaturing conditions

      • ELISA: soluble proteins with accessible surface epitopes

      • Immunohistochemistry: fixed epitopes with potential masking from fixation

      • Flow cytometry: native membrane proteins in cellular context

    • Resolution strategy:

      • Map the precise epitope using peptide arrays or hydrogen-deuterium exchange

      • Test multiple antibodies targeting different epitopes on the same protein

      • Implement epitope retrieval optimization specific to each platform

  • Sample preparation variables:

    • Methodological investigation: Systematically evaluate how sample preparation affects epitope recognition

      • Fixation methods: Compare formaldehyde, methanol, acetone effects

      • Buffer compositions: Test detergent types/concentrations and ionic strength

      • Blocking reagents: Evaluate BSA, casein, commercial blockers for background reduction

    • Resolution approach:

      • Develop platform-specific protocols optimized for each antibody

      • Document all preparation variables in standardized formats

      • Validate with known positive and negative controls under identical conditions

  • Antibody validation matrix:

    • Methodological framework: Implement a comprehensive validation strategy across platforms

      • Test multiple antibody concentrations/dilutions for each platform

      • Include genetic knockout/knockdown controls alongside wild-type samples

      • Perform peptide competition assays to confirm specificity

      • Compare multiple antibody clones against the same target

    • Documentation approach:

      • Create standardized validation reports with quantitative metrics

      • Document batch information and storage conditions

      • Maintain platform-specific positive and negative control data

  • Antibody quality assessment:

    • Methodological evaluation: Analyze physical characteristics of the antibody preparation

      • Aggregation status via SEC-HPLC or dynamic light scattering

      • Fragmentation analysis via SDS-PAGE under reducing/non-reducing conditions

      • Charge heterogeneity via isoelectric focusing or ion-exchange chromatography

    • Resolution strategy:

      • Implement size exclusion chromatography to remove aggregates

      • Optimize storage conditions (buffer, temperature, concentration)

      • Consider reformulation with stabilizing excipients

  • Cross-reactivity profiling:

    • Methodological approach: Determine if inconsistencies stem from differential cross-reactivity

      • Test against recombinant protein family members

      • Evaluate species cross-reactivity with orthologous proteins

      • Analyze potential post-translational modification recognition

    • Resolution strategy:

      • Select antibodies with appropriate cross-reactivity profiles for each application

      • Implement additional blocking steps for platforms where cross-reactivity occurs

      • Consider developing new antibodies with enhanced specificity

Implementation of this systematic troubleshooting approach can resolve up to 80-90% of inconsistency issues across experimental platforms. For persistent problems, researchers should consider developing new antibodies using different immunization strategies or in vitro display technologies targeting alternative epitopes .

How can researchers differentiate between true target binding and artifactual signals in monoclonal antibody experiments?

Differentiating between true target binding and artifactual signals is crucial for generating reliable data with monoclonal antibodies. Researchers should implement a comprehensive validation framework incorporating multiple orthogonal approaches:

  • Genetic knockout validation:

    • Gold standard methodology: Compare antibody signals between:

      • Wild-type samples expressing the target

      • Samples with genetic deletion/disruption of the target (CRISPR/Cas9, TALEN, or siRNA)

      • Samples with target overexpression as positive controls

    • Implementation approach:

      • True target binding: Complete signal loss in knockout samples

      • Artifactual binding: Persistent signal in knockout samples

      • Quantitative analysis: At least 90% signal reduction expected with true binding

  • Epitope competition assays:

    • Methodological principle: Pre-incubation with purified target protein or synthetic peptides corresponding to the epitope should competitively inhibit true binding

    • Execution strategy:

      • Titrate increasing concentrations of competing antigen (10⁻⁹ to 10⁻⁵ M)

      • Include non-relevant proteins/peptides as negative controls

      • Quantify dose-dependent signal reduction

    • Interpretation framework:

      • True binding: Specific competition with target but not control proteins

      • Artifactual signal: No competition or non-specific reduction with all proteins

  • Independent antibody correlation:

    • Methodological approach: Compare signals from multiple antibodies recognizing different epitopes on the same target

    • Implementation strategy:

      • Test at least three independent antibodies (different clones, different species)

      • Analyze signal co-localization in imaging applications

      • Compare quantitative measurements across antibodies

    • Analysis framework:

      • True binding: High correlation between signals from independent antibodies (r > 0.8)

      • Artifactual binding: Poor correlation between antibody signals

  • Orthogonal detection technologies:

    • Methodological principle: Validate antibody results using antibody-independent methods

    • Technology options:

      • Mass spectrometry for protein identification and quantification

      • RNA sequencing or qPCR to correlate protein with mRNA levels

      • Activity-based assays for functional proteins

    • Interpretation approach:

      • True binding: Concordance between antibody data and orthogonal methods

      • Artifactual signal: Significant discrepancies between methods

  • Tag-based validation systems:

    • Methodological strategy: Engineer epitope tags into endogenous loci or expression constructs

    • Implementation approach:

      • CRISPR knock-in of small epitope tags (HA, FLAG, V5)

      • Compare signals between target-specific antibody and tag-specific antibody

      • Include untagged controls to assess tag-antibody specificity

    • Analysis framework:

      • True binding: Co-localization of target and tag signals

      • Artifactual binding: Discrepancies between target and tag signals

  • Critical controls for specific artifacts:

    • For non-specific Fc receptor binding:

      • Include isotype-matched control antibodies

      • Pre-block with Fc receptor blocking reagents

      • Test F(ab')₂ fragments lacking Fc regions

    • For endogenous peroxidase/phosphatase activity:

      • Include secondary-only controls

      • Implement specific blocking steps

      • Use fluorescence-based detection as alternative

Implementation of these validation approaches should be documented in standardized formats to facilitate comparison across experiments and laboratories. A minimum of three independent validation methods should be applied before concluding that antibody signals represent true target binding .

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.