GIP Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to GIP Antibodies

Gastric Inhibitory Polypeptide (GIP) antibodies are specialized immunoglobulins designed to target GIP or its receptor (GIPR), a G protein-coupled receptor critical for metabolic regulation. GIP, an incretin hormone, stimulates insulin secretion in response to nutrient intake and modulates lipid metabolism . Antibodies against GIP/GIPR have emerged as tools for studying metabolic pathways and as therapeutic candidates for obesity, diabetes, and related disorders .

Mechanism of Action

GIP antibodies function through two primary mechanisms:

  • Neutralization: Blocking GIP binding to GIPR, thereby inhibiting downstream signaling .

  • Receptor Antagonism: Directly binding to GIPR to prevent activation, as seen with monoclonal antibodies (mAbs) like mAb2, which locks the receptor in an auto-inhibited state .

Structurally, antagonistic antibodies (e.g., mAb2) bind the GIPR extracellular domain and its C-terminal stalk, mimicking ligand interactions to suppress activity . In contrast, non-neutralizing antibodies (e.g., mAb1) bind without affecting ligand binding .

Monoclonal Antibodies

  • mAb2: A full antagonist that reduces body weight and improves glucose metabolism in preclinical models .

  • AMG 133: A bispecific antibody-conjugate combining GIPR antagonism with glucagon-like peptide-1 receptor (GLP-1R) agonism, showing synergistic weight loss in humans .

Polyclonal Antibodies

  • Proteintech 18034-1-AP: A rabbit polyclonal antibody used for immunohistochemistry (IHC) and ELISA, reactive across human, mouse, and rat tissues .

  • Synaptic Systems 514 003: Targets mouse/rat GIP, with applications in IHC and Western blotting .

Preclinical Studies

  • Weight Loss: Central GIPR inhibition in mice reduced body weight, fat accumulation, and hypothalamic inflammation .

  • Metabolic Improvements: GIPR antagonist antibodies improved insulin sensitivity and reduced proinflammatory cytokines (e.g., IL-6) in adipose tissue .

  • Bispecific Molecules: Conjugating GIPR-Ab to GLP-1 peptides amplified cAMP signaling in cells expressing both receptors, enhancing weight loss in diet-induced obesity (DIO) mice .

Table 2: Efficacy of AMG 133 in Preclinical Models

ModelDoseOutcomeReference
DIO Mice10 mg/kg30% body weight reduction over 6 weeks
Cynomolgus Monkeys3 mg/kgSustained weight loss for 150 days

Clinical Trials

  • Phase 1 Study (AMG 133): A single subcutaneous dose (21–420 mg) in obese patients resulted in dose-dependent weight loss (up to 14.5%) with no severe adverse events .

Therapeutic Applications

  • Obesity: GIPR antagonism reduces adiposity and enhances leptin sensitivity .

  • Diabetes: Combining GIPR-Ab with GLP-1R agonists improves glycemic control beyond GLP-1 monotherapy .

  • Neuroprotection: GIP antibodies may mitigate neuroinflammation, as GIPR is expressed in brain regions linked to energy homeostasis .

Challenges and Future Directions

  • Species Specificity: Some antibodies (e.g., AMG 133) show limited cross-reactivity with rodent GIPR, necessitating surrogate molecules for murine studies .

  • Long-Term Safety: Prolonged GIPR blockade may require monitoring for unintended effects on bone metabolism or cardiovascular health .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
Gastric Inhibitory Peptide antibody; Gastric inhibitory polypeptide antibody; Gastric inhibitory polypeptide precursor antibody; GIP antibody; GIP_HUMAN antibody; Glucose dependent insulinotropic polypeptide antibody; Glucose-dependent insulinotropic polypeptide antibody; Incretin hormone antibody
Target Names
GIP
Uniprot No.

Target Background

Function
GIP is a potent stimulator of insulin secretion and exhibits relatively weak inhibition of gastric acid secretion.
Gene References Into Functions
  1. Genetic variability in the GIP gene has been linked to coronary artery disease, potentially contributing to premature coronary artery disease in Chinese Han individuals with type 2 diabetes. PMID: 29765988
  2. A truncated form of GIP, GIP(3-30)NH2, has been shown to antagonize the physiological effects of GIP in glucose metabolism, subcutaneous abdominal adipose tissue blood flow, and lipid metabolism in humans. PMID: 28667118
  3. Plasma concentrations of GIP and PP are lower in pancreatic cancer patients, regardless of the degree of glucose intolerance, compared to Type 2 diabetic patients and healthy controls. PMID: 28027898
  4. Evening postprandial insulin and GIP responses, along with insulin resistance, decreased by over 30% after three meals limiting daily carbohydrate intake to 30%, compared to no such changes after three 60%-carbohydrate meals. This effect was independent of pre-meal exercise. PMID: 27798656
  5. The stimulatory effect of IGF-1 on the GIP promoter supports the hypothesis of a functional growth hormone-IGF-1-GIP axis. PMID: 28179449
  6. Lower maternal 25OHD levels may be associated with decreased cord 25OHD and increased cord GLP-1 and GIP levels, potentially playing a role in the transfer of maternal glucose to the fetus. PMID: 26650343
  7. Excess androgen activity might contribute to altered incretin secretion in lean women with polycystic ovary syndrome (PCOS). However, it cannot be ruled out that increased GIP levels might also induce hyperandrogenemia in PCOS. PMID: 26895276
  8. Our findings suggest an altered DPP4-incretin system and altered immunoregulation, including potentially dysfunctional GLP1(9)(-)(36) signaling in type 1 diabetes. PMID: 26434625
  9. Fasting GIP concentrations are higher in individuals with a history of cardiovascular disease (myocardial infarction, stroke) compared to control subjects. PMID: 26395740
  10. Data suggest that high levels of blood glucose or AGEs (advanced glycation end products), as observed in hyperglycemia, reduce insulin secretion by pancreatic beta cells through antagonism of GIP (gastric inhibitory polypeptide)/GIP receptor signaling. PMID: 26221611
  11. Postprandial plasma levels of glucose-dependent insulinotropic polypeptide (GIP) and insulin (INS) are responsive to the glycemic index of consumed foods. The glycemic index of breakfast cereals regulates plasma postprandial GIP and INS. PMID: 25852025
  12. Irisin and GIP may contribute to the development of polycystic ovary syndrome and might also represent novel biomarkers for the condition. PMID: 25029417
  13. Dietary interventions can regulate postprandial blood levels of both GIP and insulin. Inclusion of nopal/Opuntia/cactus (a functional food in traditional Mexican medicine) in breakfast reduces postprandial levels of GIP and insulin. PMID: 25132122
  14. Phosphatidylinositol 3-kinase gamma plays a role in insulin secretion induced by glucose-dependent insulinotropic polypeptide. PMID: 25288806
  15. These findings support the notion that the GIP-GIPR axis plays a role in the etiology of central obesity in humans. PMID: 25324507
  16. Studies in healthy Japanese men suggest that plasma GIP levels in the postprandial period increase dose-dependently with fat content of meals of ordinary size, even with relatively small additions of fat. PMID: 24507870
  17. Patients with idiopathic gastroparesis exhibit abnormal GIP levels. PMID: 23663508
  18. Beta cell connectedness is an inherent property of human islets that is likely to influence incretin-potentiated insulin secretion. PMID: 24018562
  19. Data suggest that postprandial plasma levels of glucose-dependent insulinotropic peptide (GIP) and glucagon-like peptide-1 (GLP1) are increased after consumption of buckwheat crackers compared to rice crackers in healthy and type 2 diabetic subjects. PMID: 23485142
  20. GIP induces an inflammatory and prolipolytic response via the PKA -NF-kappaB-IL-1 pathway and impairs insulin sensitivity of glucose uptake in human adipocytes. PMID: 23092914
  21. Results indicate that postprandial GIP secretion in the early phase after a test meal in Japanese patients with type 2 diabetes was positively correlated with BMI, but not in those with type 1 diabetes. PMID: 22301939
  22. Subjects with metabolic syndrome and hyperinsulinemia showed increased GIP secretion, which could be responsible for delayed glucagon suppression. PMID: 22391044
  23. Reduced insulinotropic effects of GIP or GLP-1 (as seen in type 2 diabetes) can be induced in healthy subjects. This indicates that reduced incretin stimulation of insulin secretion results from insulin resistance/glucose intolerance. PMID: 22319034
  24. GIP reduces free fatty acid release from adipose tissue by inhibiting lipolysis or by increased reesterification. PMID: 22179810
  25. GIP may have a pro-obesogenic action [review]. PMID: 21815989
  26. Studies have identified some potentially important additional C-terminal interactions of GIP with its N-terminal extracellular receptor domain. PMID: 21539943
  27. The human GIP locus was differentially selected in East Asians about 8100 years ago based on the analysis of a nonsynonymous SNP (rs2291725). PMID: 20978139
  28. GLP-2, but not GIP, was found to stimulate glucagon release in patients with type 1 diabetes, suggesting a role for GLP-2 in the postprandial hyperglucagonaemia characteristic of individuals with type 1 diabetes. PMID: 20580750
  29. These results demonstrate that Tyr/His(1) and Ile/Thr(7) of GIP/GLP-1 peptides confer differential ligand selectivity toward GIPR and GLP1R. PMID: 20799012
  30. This research demonstrates for the first time that changes in insulin secretion after lifestyle interventions may be mediated through alterations in GIP secretion from intestinal K-cells. PMID: 20200305
  31. No statistically significant association was observed between any of the single nucleotide polymorphisms of GIP analyzed and type 2 diabetes in the studied population. PMID: 20673334
  32. GIP is expressed in and secreted from pancreatic islets, promoting islet glucose competence and potentially supporting islet development and/or survival. PMID: 20138041
  33. A binding mode of GIP to GIPR has been identified in which the N-terminal moiety of GIP is situated within transmembrane helices (TMH) 2, 3, 5, and 6, with biologically crucial Tyr1 interacting with Gln224 (TMH3), Arg300 (TMH5), and Phe357 (TMH6). PMID: 20061446
  34. Substitution of Glu(3) in GIP with proline produces a novel dipeptidylpeptidase IV-resistant GIP antagonist that inhibits GIP-induced cAMP generation and insulin secretion with high sensitivity and specificity in vitro. PMID: 11820780
  35. GIP activates the Raf-Mek1/2-ERK1/2 module through a cyclic AMP/cAMP-dependent protein kinase/Rap1-mediated pathway. PMID: 12138104
  36. Mutations in the promoter region of the GIP receptor gene are unlikely to underlie GIP-dependent Cushing syndrome. PMID: 12530694
  37. Elevated plasma GIP levels are correlated with hyperinsulinemia in the impaired glucose-tolerant state, while type 2 diabetes is associated with a failure to secrete adequate amounts of GIP. PMID: 15220248
  38. Bombesin and nutrients additively stimulate GIP release from GIP/Ins cells. PMID: 15383372
  39. This research describes the solution structure of GIP(1-30)amide, the major biologically active fragment of glucose-dependent insulinotropic polypeptide. PMID: 15522230
  40. GIP augments glucose-stimulated insulin secretion and acts as an endogenous inhibitor of gastric acid secretion--REVIEW. PMID: 15533777
  41. GIP stimulates insulin secretion by potentiating events underlying membrane depolarization and exerting direct effects on exocytosis. PMID: 15955806
  42. The relationship between insulin resistance and the insulin secretion to GIP suggests that beta cell secretory function in response to different stimuli increases adaptively when insulin sensitivity is diminished, as in gestational diabetes. PMID: 16010522
  43. GIP is rapidly degraded into inactive metabolites by the enzyme dipeptidyl-peptidase-IV (review). PMID: 16142014
  44. Protein kinase B, LKB1, and AMP-activated protein kinase have roles in the activation of lipoprotein lipase by glucose-dependent insulinotropic polypeptide in adipocytes. PMID: 17244606
  45. This study identified a splice site mutation of the Glucose-dependent insulinotropic polypeptide (GIP) gene, resulting in a truncated protein and providing evidence for an association between GIP receptor genotype and cardiovascular disease. PMID: 17624916
  46. GIP has a physiological role in lipid homeostasis and potentially in the pathogenesis of obesity. PMID: 18054552
  47. Concomitant expression of Pax6 and Pdx1 is important for glucose-dependent insulinotropic polypeptide expression. PMID: 18593849
  48. GIP secretion is blunted after the biliopancreatic diversion only in diabetic patients, suggesting a role in insulin resistance and diabetes. PMID: 19229515
  49. GIP may mediate the attenuated glucose-stimulated insulin response after exercise/diet interventions. PMID: 19351807
  50. Inhibition of apoptosis by GIP is mediated via a key pathway involving Akt-dependent inhibition of apoptosis signal-regulating kinase 1, which subsequently prevents the pro-apoptotic actions of p38 MAPK and JNK. PMID: 19748889

Show More

Hide All

Database Links

HGNC: 4270

OMIM: 137240

KEGG: hsa:2695

STRING: 9606.ENSP00000350005

UniGene: Hs.1454

Protein Families
Glucagon family
Subcellular Location
Secreted.

Q&A

What is GIP and why are antibodies against it important in research?

GIP (glucose-dependent insulinotropic polypeptide, also known as gastric inhibitory polypeptide) is an incretin hormone that plays crucial roles in glucose homeostasis and energy metabolism. In humans, GIP is a 153 amino acid secreted protein with a mass of approximately 17.1 kDa. It belongs to the Glucagon protein family and functions as a potent stimulator of insulin secretion and a relatively poor inhibitor of gastric acid secretion .

GIP antibodies are essential research tools because:

  • They enable specific detection and quantification of GIP in various biological samples

  • They allow researchers to study GIP's role in metabolic pathways

  • They can be developed as antagonists to block GIP signaling for investigating metabolic disorders

  • They provide means to explore GIP's involvement in obesity and diabetes pathophysiology

GIP also regulates fat metabolism by acting on adipocytes, thus playing a significant role in energy homeostasis . Understanding these functions requires specific tools like antibodies that can detect, quantify, or modulate GIP activity.

What are the primary differences between GIP antibodies and GIPR antibodies?

GIP antibodies target the hormone itself, while GIPR antibodies target the GIP receptor. This distinction is critical for research design:

GIP Antibodies:

  • Bind directly to GIP hormone

  • Primarily used for detection and quantification (ELISA, IHC, WB)

  • Can neutralize circulating GIP

  • Applications include measuring GIP levels in plasma and tissue samples

GIPR Antibodies:

  • Bind to the GIP receptor (GIPR)

  • Can function as antagonists (blocking receptor) or non-neutralizing binders

  • Used to study receptor distribution and function

  • Can be developed as therapeutic agents for metabolic diseases

  • Can be designed as bispecific molecules when combined with other peptides

Research shows that GIPR antagonist antibodies can reduce body weight and improve metabolic parameters in animal models, indicating their potential as therapeutic agents for treating obesity and related disorders .

How should researchers select the appropriate GIP antibody for their experimental applications?

Selection of the appropriate GIP antibody requires careful consideration of several factors:

Key Selection Criteria:

ParameterConsiderations
SpecificityVerify cross-reactivity with relevant species (human, mouse, rat, etc.)
ApplicationMatch antibody to technique (WB, ELISA, IHC, IF, neutralization)
EpitopeChoose antibodies targeting relevant domains of GIP/GIPR
FormatSelect appropriate format (unconjugated, biotinylated, fluorescent-tagged)
ValidationReview published validation data and citations
Clone typeConsider monoclonal vs. polyclonal based on needs

For instance, search results show various commercial antibodies with different applications - some optimized for WB, ELISA, and IHC . When studying receptor antagonism, researchers should select antibodies specifically validated for neutralization assays, such as those that have demonstrated antagonistic activity in cAMP assays .

For structural studies, antibodies that have been successfully used in crystallography might be preferred, as seen with antibodies like mAb1 and mAb2 that have been co-crystallized with GIPR .

What assays can be used to validate GIP antibody specificity and function?

Several complementary assays should be employed to thoroughly validate GIP antibodies:

For Binding Specificity:

  • ELISA to determine binding affinity (KD) values

  • KinExA assay for precise affinity measurements (as used for mAb1 and mAb2 with KD values of 144 pM and 68 pM respectively)

  • Western blotting against recombinant GIP and tissue lysates

  • Immunohistochemistry on tissues known to express GIP (e.g., duodenum, small intestine)

  • Receptor binding assays using fluorescent microvolume assay technology (FMAT)

For Functional Assessment:

  • cAMP assays to measure GIP-stimulated cAMP production and antibody inhibition

  • Cell-based functional assays (IC50 determination)

  • Receptor ligand competition assays using labeled GIP peptide

  • Schild analysis to determine the mechanism of antagonism

For GIPR antagonist antibodies, researchers should perform dose-response experiments to determine IC50 values and maximum inhibition percentages. For example, mAb2 was characterized as a full antagonist (IC50 of 48 nM) while mAb3 and mAb4 were partial antagonists (IC50 of 1.7 nM and 3.1 nM) with maximal inhibition of 29% and 50%, respectively .

How can researchers accurately measure GIP responses using antibody-based methods?

Measuring GIP responses requires careful consideration of antibody selection and assay design:

Methodological Considerations:

  • Antibody selection is critical: Different antibodies can yield significantly different results. For example, one study using three different antibodies (S100, GP01, and GP24) to measure GIP responses in diabetes found three to sixfold higher levels of GIP when using GP24 compared to the other antibodies .

  • Sample preparation: For plasma samples, alcohol extraction is recommended before assaying, as demonstrated in studies measuring GIP responses in diabetic patients .

  • Assay validation: Researchers should validate their assays using appropriate controls:

    • Recombinant GIP standards

    • Samples from GIP knockout models (negative control)

    • Samples with known GIP concentrations

  • Cross-reactivity evaluation: Test for potential cross-reactivity with related peptides (GLP-1, glucagon) to ensure specificity.

  • Standardization: Use standardized protocols and calibrators to allow for comparison between studies.

The contradictory reports of GIP responses in diabetes using different radioimmunoassay systems highlight the importance of antibody selection . Researchers should be aware that different antibody systems might recognize different forms of GIP (e.g., 5 kDa and 8 kDa forms) or structurally related peptides, potentially leading to conflicting results.

How are GIPR antagonist antibodies being developed as potential therapeutic agents?

GIPR antagonist antibodies represent a promising approach for treating obesity and metabolic disorders, with research revealing several key development strategies:

Therapeutic Development Approaches:

  • Monoclonal antibody development:

    • Phage display libraries have successfully yielded potent GIPR antagonists like Gipg013

    • Chicken immunization has proven particularly effective, generating diverse panels of antagonistic antibodies where mammalian hosts failed due to high conservation of GIPR

  • Bispecific molecule engineering:

    • GIPR antagonist antibodies conjugated to GLP-1 receptor agonists show enhanced efficacy

    • GIPR-Ab/GLP-1 bispecific molecules demonstrate synergistic effects on body weight reduction in DIO mice and monkeys

    • Chemical conjugation of GLP-1 peptides to site-specific engineered cysteines (E384C) on hGIPR-Ab yields functional bispecific molecules

  • Mechanism of action characterization:

    • Antagonistic antibodies like mAb2 not only occlude ligand peptide binding but also recognize the GIPR C-terminal stalk region, locking GIPR in a novel auto-inhibited state

    • Crystal structures of GIPR ECD in complex with antagonistic antibodies reveal binding to the N-terminal α-helix and the conserved glucagon receptor subfamily recognition fold

Administration of antagonistic GIPR antibodies in diet-induced obesity mice for 7 weeks leads to both reduction in body weight gain and improvement of metabolic profiles, supporting their therapeutic potential .

What molecular mechanisms explain the efficacy of GIPR antagonism for metabolic disorders?

The molecular basis for GIPR antagonism's efficacy involves several intricate mechanisms:

  • Genetic foundations:

    • Genome-wide association studies have identified variants with reduced activity at the human GIPR locus associated with reduced BMI

    • Genetic ablation of GIPR leads to decreased body weight in diet-induced obese mice

  • Competitive antagonism mechanisms:

    • Structural studies show antagonistic antibodies bind through hydrogen bonds to the N-terminal α-helix of GIPR ECD and the conserved glucagon receptor subfamily recognition fold

    • The antibody epitope overlaps with the GIP binding site, ensuring competitive antagonism

  • Novel auto-inhibition mechanism:

    • Antagonistic antibodies like mAb2 not only partially occlude ligand peptide binding but also recognize the GIPR C-terminal stalk region in a helical conformation

    • This acts as a molecular mimic of the ligand peptide and locks GIPR in a novel auto-inhibited state

  • Adipocyte-specific effects:

    • GIPR activity in adipocytes is partially responsible for the ability of GIPR antagonists to prevent weight gain in DIO mice

    • This demonstrates a role of adipocyte GIPR in the regulation of adiposity in vivo

Interestingly, both GIPR antagonism and agonism show similar preclinical body weight effects. This paradox is explained by chronic GIPR agonism desensitizing GIPR activity in primary adipocytes (both differentiated in vitro and in adipose tissue in vivo), effectively functioning like a GIPR antagonist .

How does the combination of GIPR antagonism with GLP-1R agonism enhance therapeutic potential?

The combination of GIPR antagonism with GLP-1R agonism represents a breakthrough approach for treating obesity and metabolic disorders:

Synergistic Effects:

  • Enhanced body weight reduction:

    • GIPR-Ab/GLP-1 bispecific molecules produce greater weight loss than either GIPR-Ab or control antibody conjugates alone

    • This synergistic effect has been demonstrated in both DIO mice and monkeys

  • Metabolic improvements:

    • The combination improves multiple metabolic parameters beyond what is achieved with single-target approaches

    • GIPR-Ab/GLP-1 reduces the respiratory exchange ratio in DIO mice, indicating altered substrate utilization

  • Molecular mechanism of synergy:

    • Simultaneous receptor binding and rapid receptor internalization by GIPR-Ab/GLP-1 amplify endosomal cAMP production in recombinant cells expressing both receptors

    • This phenomenon may explain the enhanced efficacy of the bispecific molecules

  • Design considerations:

    • Bispecific molecules are created by conjugating GLP-1 peptides containing amino-acid modifications (to extend half-life while optimizing potency) to GIPR antagonist antibodies

    • The conjugation site (e.g., E384C) is critical for maintaining antagonist activity and achieving favorable pharmacokinetic profiles

Pharmacokinetic studies show that labeled mGIPR-Ab and mGIPR-Ab/P1 have similar serum concentration-time profiles, indicating that the conjugation of GLP-1 peptide does not significantly affect the antibody's circulation time .

Why might different GIP antibodies yield contradictory experimental results?

Contradictory results when using different GIP antibodies can arise from several factors:

Potential Sources of Discrepancy:

  • Epitope specificity:

    • Different antibodies recognize distinct epitopes on GIP or GIPR

    • Some antibodies may detect only specific forms or fragments of GIP

    • For example, studies using three different antibodies (S100, GP01, and GP24) showed dramatically different GIP measurements in diabetic patients

  • Recognition of GIP isoforms:

    • Structurally abnormal variable cross-reacting 5000 dalton (5 kDa) and 8 kDa GIP forms may be recognized differently by various antibodies

    • Unidentified structurally GIP-related peptides associated with type 2 diabetes might contribute to conflicting results

  • Functional differences:

    • Some antibodies are full antagonists while others are partial antagonists

    • For example, mAb2 completely reverses GIP-induced cAMP production, while mAb3 and mAb4 only partially antagonize GIP activity (29% and 50% maximal inhibition, respectively)

  • Technical variation:

    • Differences in assay conditions, sample preparation, and detection methods

    • Variation in antibody performance across different applications (ELISA vs. IHC vs. WB)

To address these issues, researchers should:

  • Use multiple antibodies targeting different epitopes when possible

  • Include appropriate positive and negative controls

  • Validate antibodies in their specific experimental system

  • Consider complementary non-antibody-based approaches to confirm findings

How should species differences be considered when selecting and using GIP antibodies?

Species differences significantly impact GIP antibody selection and experimental design:

Cross-Reactivity Considerations:

  • Conservation challenges:

    • GIPR is highly conserved between rodents and humans, which has contributed to previous mouse and rat immunization campaigns generating very few usable antibodies

    • This conservation makes it difficult to generate antibodies that recognize species-specific differences

  • Alternative immunization strategies:

    • Chicken immunization has emerged as a valuable approach for generating diverse antibody panels

    • Chickens, being phylogenetically distant from mammals, can generate antibodies against conserved mammalian proteins

    • One chicken immunization campaign generated 172 unique GIPR antibody sequences, with three-quarters functioning as antagonists

  • Documented cross-reactivity:

    • Researchers should verify antibody cross-reactivity with the specific species of interest

    • Some antibodies are species-specific while others show broad cross-reactivity

    • For example, hGIPR-Ab shows comparable antagonist activity in cells expressing human GIPR (IC50 = 103.6 nM) and monkey GIPR (IC50 = 24.6 nM)

  • Testing protocols:

    • Species-specific validation can be performed using:

      • Recombinant GIP/GIPR from different species

      • Cell lines expressing species-specific receptors

      • Tissues from different animal models

    • For instance, stably transfected cell lines overexpressing human, dog, mouse, or rat receptors were used to evaluate antibody binding specificity in receptor ligand binding assays

When designing experiments spanning multiple species, researchers should select antibodies validated for cross-reactivity or use species-specific antibodies as appropriate.

What controls and validation steps are essential when characterizing novel GIP antibodies?

Rigorous characterization of novel GIP antibodies requires comprehensive controls and validation:

Essential Validation Protocol:

  • Binding affinity determination:

    • KinExA assay to measure precise KD values

    • ELISA to confirm antigen recognition

    • Surface plasmon resonance or biolayer interferometry for real-time binding kinetics

  • Specificity controls:

    • Testing against related peptides (GLP-1, glucagon)

    • Using GIP/GIPR knockout models or cells

    • Competitive binding with known ligands

  • Functional characterization:

    • cAMP assays using precise concentrations of GIP (e.g., 6 pM mouse GIP as described for testing mAb1 and mAb2)

    • Schild analysis to determine antagonist potency and mechanism

    • Dose-response curves at different antibody concentrations

    • Controls using non-binding isotype-matched antibodies (e.g., NIP228 as control for Gipg013)

  • Structural validation:

    • Epitope mapping through mutagenesis or structural studies

    • X-ray crystallography to determine binding interfaces (as performed for mAb1 and mAb2)

    • Hydrogen-deuterium exchange mass spectrometry

  • In vivo validation:

    • Pharmacokinetic profiling (e.g., using 111In-DOTA-labeled antibodies)

    • Assessment of body weight and metabolic parameters in appropriate animal models

    • Comparison with established GIPR modulators

For example, a comprehensive characterization of mAb2 included KinExA for binding (KD = 68 pM), cAMP assays for antagonism (IC50 = 48 nM), structural studies to determine the binding interface, and in vivo studies showing reduced body weight gain in DIO mice after 7 weeks of treatment .

How are bispecific antibody approaches advancing GIP research beyond traditional monoclonal antibodies?

Bispecific antibody approaches represent a significant advancement in GIP research with novel capabilities:

Bispecific Innovation:

  • GIPR-Ab/GLP-1 design strategies:

    • Chemical conjugation of GLP-1 peptides to site-specific engineered cysteines (E384C) on GIPR antibodies

    • Optimization of linker length and composition ((GGGGS)3) for optimal dual-targeting

    • Peptide modifications to extend half-life while preserving potency

  • Mechanisms of enhanced efficacy:

    • Simultaneous receptor binding triggers unique signaling patterns

    • Co-internalization of both receptors enhances endosomal cAMP production

    • Synergistic effects on metabolic parameters beyond additive effects of individual components

  • Comparative advantages:

    • Greater weight loss than individual therapies or combinations of separate agents

    • Improved dosing convenience compared to co-administration approaches

    • Potential for tailored receptor engagement by modifying peptide potency (e.g., P1 vs P2 versions)

  • Technical considerations:

    • Conjugation site selection is critical for maintaining antagonist activity

    • The E384C site was selected based on favorable alkylation efficiency and PK profile compared to other sites

    • Antibody function preservation after conjugation must be verified (e.g., hGIPR-Ab/P1 and hGIPR-Ab/P2 maintained antagonist activity comparable to unconjugated hGIPR-Ab)

This approach has demonstrated superior efficacy in animal models and represents a promising direction for developing more effective metabolic disease therapies.

What structural insights have advanced our understanding of GIP antibody mechanisms of action?

Crystal structures of antibody-GIPR complexes have provided critical insights into antagonism mechanisms:

Structural Revelations:

  • Binding interfaces:

    • Non-neutralizing antibody (mAb1) binds GIPR without competing with the ligand peptide

    • Antagonistic antibody (mAb2) partially occludes the ligand peptide binding site

    • mAb2 additionally recognizes the GIPR C-terminal stalk region in a helical conformation

  • Novel auto-inhibition mechanism:

    • The C-terminal stalk recognition by mAb2 acts as a molecular mimic of the ligand peptide

    • This locks GIPR in a novel auto-inhibited state, explaining its superior antagonistic properties

  • Epitope mapping:

    • Gipg013 binds through hydrogen bonds from its complementarity-determining regions to the N-terminal α-helix of GIPR ECD

    • Additional bonds form with residues around the highly conserved glucagon receptor subfamily recognition fold

    • This binding pattern overlaps with the GIP binding site, ensuring competitive antagonism

  • Resolution details:

    • Co-crystal structures of GIPR ECD with mAb1 and mAb2 were determined at resolutions of 2.1 and 2.6 Å, respectively

    • The Gipg013 Fab-GIPR ECD structure was solved by molecular replacement using polyalanine models of incretin-bound extracellular domain (PDB code 2QKH) and murine IGG1 λ antibody (PDB code 1GIG)

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.