GPNMB Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to GPNMB Recombinant Monoclonal Antibody

GPNMB (Glycoprotein Non-Metastatic Melanoma Protein B), also known as osteoactivin or DC-HIL, is a transmembrane glycoprotein overexpressed in cancers such as glioblastoma, melanoma, and triple-negative breast cancer. Recombinant monoclonal antibodies targeting GPNMB are engineered to bind with high specificity to its extracellular domain, enabling therapeutic applications like immunotoxins, antibody-drug conjugates (ADCs), and diagnostic tools . These antibodies are produced using advanced technologies such as phage display libraries and yeast surface display systems to optimize affinity and functionality .

Development and Engineering

Recombinant monoclonal anti-GPNMB antibodies are developed through:

  • Phage Display Libraries: Isolation of single-chain variable fragments (scFvs) with subsequent affinity maturation via random mutagenesis (e.g., G49 scFv evolved into F6V-PE38 immunotoxin with 28-fold improved affinity) .

  • Yeast Surface Display: Enables screening for clones with enhanced binding kinetics .

  • Humanized Formats: Minimize immunogenicity while retaining high specificity (e.g., CR011, a fully human monoclonal antibody) .

Key Engineering Milestones

Antibody CloneAffinity ImprovementApplication
G49 (parent)BaselineImmunotoxin
F6V28-fold increaseCancer therapy
CR011N/AADC (CR011-vcMMAE)

Immunotoxins and ADCs

  • F6V-PE38: A recombinant immunotoxin combining the F6V scFv with Pseudomonas exotoxin A. Demonstrates potent cytotoxicity in GPNMB-positive glioma and melanoma cells (IC<sub>50</sub> = 0.5 ng/mL) .

  • CR011-vcMMAE: An ADC linking CR011 to monomethyl auristatin E (MMAE). Achieves complete tumor regression in melanoma xenografts at 1.25 mg/kg .

Mechanisms of Action

  • Targeted Cytotoxicity: Internalization of ADCs delivers toxins directly to cancer cells .

  • Immune Modulation: Binds to α5β1 integrin to inhibit metastasis and enhance chemosensitivity in breast cancer .

Prognostic Value

  • High GPNMB expression correlates with poor survival in glioblastoma (HR = 3.0) and triple-negative breast cancer .

  • Surface density ranges from 1.1–7.8 × 10<sup>4</sup> molecules/cell in glioblastoma, enabling effective targeting .

Preclinical Efficacy

ModelAntibodyOutcome
Glioma xenograftsF6V-PE38Tumor regression (60% reduction)
Melanoma meningitisF6V-PE38Prolonged survival in rats
Breast cancerCDX-011 (ADC)Reduced metastasis

Research Findings and Validation

  • Binding Kinetics: Antibodies like G203 and F105 exhibit affinities of 2.7 × 10<sup>-8</sup> M and 1.6 × 10<sup>-8</sup> M, respectively .

  • Cross-Reactivity: Clone 66926-1-Ig reacts with human, mouse, and rat GPNMB, enabling translational studies .

  • Functional Assays: Antibodies inhibit VEGF signaling and reduce tumor growth in vivo via integrin-mediated pathways .

Future Directions

  • Clinical Trials: Evaluation of CR011-vcMMAE in metastatic melanoma and CDX-011 in triple-negative breast cancer .

  • Companion Diagnostics: Development of GPNMB-targeted antibodies for immunohistochemical stratification of patients .

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 the products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method or location. For specific delivery timelines, please consult your local distributors.
Synonyms
Glycoprotein (transmembrane) nmb antibody; Glycoprotein nmb antibody; Glycoprotein nmb like protein antibody; Gpnmb antibody; GPNMB_HUMAN antibody; HGFIN antibody; NMB antibody; Osteoactivin antibody; Transmembrane glycoprotein antibody; Transmembrane glycoprotein HGFIN antibody; Transmembrane glycoprotein NMB antibody
Target Names
Uniprot No.

Target Background

Function
GPNMB (Glycoprotein non-metastatic melanoma protein B) is a type I transmembrane protein that plays a role in various cellular processes, including melanosome formation, autophagy, phagocytosis, tissue repair, and negative regulation of inflammation. It has been implicated in the development and progression of various diseases, such as cancer, neurodegenerative disorders, and autoimmune diseases. Its precise function remains under investigation, but it may act as a melanogenic enzyme.
Gene References Into Functions
  1. Loss of GPNMB, a protein involved in melanosome formation, autophagy, phagocytosis, tissue repair, and inflammation regulation, is linked to autosomal-recessive Amyloidosis cutis dyschromica. PMID: 29336782
  2. Research indicates high GPNMB mRNA levels in osteosarcoma tissues and cell lines. Its silencing regulates osteosarcoma cell proliferation and metastasis by suppressing the PI3K/Akt/mTOR signaling pathway. PMID: 29620278
  3. Serum levels of GPNMB strongly correlate with the accumulation of bioactive lipid substrate of Gaucher disease, glucosylsphingosine, as well as established biomarkers, chitotriosidase and chemokine, CCL18. PMID: 28003098
  4. Increased brain expression of GPNMB is associated with a significant risk for Parkinson's disease. PMID: 28391543
  5. Studies show that GPNMB aggregates are localized to neurons, but not to astrocytes or microglia, in the spinal cord of ALS patients. GPNMB exhibits protective effects against mutant TDP-43-induced motor neuron cell death through the ERK1/2 and Akt pathways. PMID: 27935101
  6. This review summarizes the latest understanding of Gpnmb in the context of diagnosis, progression, and prognosis of pathological disorders and neoplasms, highlighting clinical advancements in targeting Gpnmb-expressing malignancies. PMID: 29441921
  7. Combining MAPK pathway inhibitors with an antibody-drug conjugate targeting GPNMB offers an effective therapeutic approach. PMID: 27515299
  8. High GPNMB expression is associated with increased invasive behavior in lung cancer. PMID: 26883195
  9. Research suggests that MAFK and its target gene GPNMB play significant roles in the malignant progression of triple-negative breast cancer (TNBC) cells, presenting potential new therapeutic targets for TNBC patients. PMID: 28400538
  10. Glycoprotein non-metastatic melanoma protein B (GPNMB) demonstrates increased expression in breast cancer in vivo compared to normal breast tissue. PMID: 28689015
  11. Osteoactivin differentially regulates the expression of matrix metalloproteases in head and neck squamous cell carcinomas to promote cancer cell invasion. PMID: 28295306
  12. GPNMB is expressed in a temporal manner during eosinophil development and delivers a proliferative signal upon activation. PMID: 28104809
  13. It may be premature to conclude associations between GPNMB rs156429 and Parkinson's disease, amyotrophic lateral sclerosis, and multiple system atrophy. PMID: 27132081
  14. GPNMB protein is highly expressed in bladder cancer, correlating with poor prognosis in bladder cancer patients. GPNMB promotes proliferation, migration, and invasion in bladder cancer cells. PMID: 28443476
  15. GPNMB acts as an inducer for glioma and enhances matrix metalloproteinase activity through the Wnt/beta-catenin pathway, contributing to glioma tumorigenesis. PMID: 27334625
  16. Findings indicate that GPNMB promotes glioma growth via Na(+)/K(+)-ATPase alpha subunits, suggesting this interaction as a potential therapeutic target for treating brain glioblastomas. PMID: 27836549
  17. GPNMB, a melanocytic marker, is upregulated in Tsc2-null mouse uteri and human lymphangioleiomyomatosis samples. PMID: 26880751
  18. GPNMB holds promise as a biomarker and therapeutic target for the development and progression of Nonalcoholic fatty liver disease in obesity. PMID: 26581806
  19. Osteoactivin in the extracellular matrix promotes oral squamous carcinoma cell adhesion and migration. PMID: 26636434
  20. GPNMB serves as a potential marker for the visceral pathology in Niemann-Pick Type C Disease. PMID: 26771826
  21. GPNMB might function as a surrogate marker for breast cancer and may interact with the HER2 signaling pathway. PMID: 26077887
  22. GPNMB plays crucial roles in regulating the expression of key pluripotency genes in dental pulp cells and modifying odontogenic differentiation. PMID: 26261527
  23. A positive correlation exists between GPNMB and NRP-1 levels in human breast tumors. PMID: 25772243
  24. Data indicates that the expression of Gpnmb and Spp1 is significantly upregulated in glioma-associated microglia/macrophages, highlighting the importance of macrophages and microglia as therapeutic targets in anti-tumor treatment regimens. PMID: 25658639
  25. GPNMB/OA protein expression prevents cells from apoptosis, enhances proliferation, and represents a novel modulator of invasion and metastasis in pancreatic cancer cells. PMID: 25426614
  26. The transcription factor MITF is a crucial regulator of GPNMB expression in dendritic cells. PMID: 25889792
  27. GPNMB mRNA in FLCN-related renal cell carcinomas was 23-fold more abundant than in sporadic tumors. PMID: 25594584
  28. GPNMB exhibits a protective effect against ischemia-reperfusion injury through phosphorylation of ERK1/2 and Akt. PMID: 25010402
  29. Glycoprotein nonmetastatic melanoma protein B plays a significant role in angiogenesis during hyperoxia injury. PMID: 25054912
  30. DC-HIL+ CD14+ HLA-DR no/low cells serve as a potential blood marker and therapeutic target for melanoma. PMID: 24933321
  31. Research identifies Gpnmb as a novel marker for obesity-induced adipose tissue macrophage infiltration, a potentiator of interleukin-4 responses, and highlights a crucial role for MITF in driving part of the adipose tissue macrophage phenotype. PMID: 24789918
  32. GPNMB/OA acts as a critical molecular mediator promoting the acquisition of a more aggressive, pro-metastatic phenotype characteristic of human DU145 and PC3 cell lines. PMID: 24589892
  33. Findings provide a new explanation for how GPNMB induces bone repair and offer a potential target for bone regeneration therapeutics and bone engineering. PMID: 23794283
  34. The PKD domain is responsible for the distinct trafficking and morphogenetic properties of PMEL and GPNMB. PMID: 23452376
  35. GPNMB expression is regulated by EpCAM and CSF-1, partially through their common downstream product c-myc. PMID: 23924854
  36. Silencing of GPNMB by siRNA inhibits melanosome formation in melanocytes in a MITF-independent manner. PMID: 22912767
  37. GPNMB inhibits motor neuron death and plays a critical role in motor neuron survival. PMID: 22891158
  38. These findings suggest that GPNMB gene is a p53- and androgen-dysregulated gene and should be considered an anti-tumor gene for prostate cancer. PMID: 22290289
  39. The abnormal expression of GPNMB may play a significant role in the development of prostate cancer. PMID: 21844952
  40. Gastrointestinal stromal tumors do not exhibit immunopositivity for ERa or HMB45. PMID: 22014058
  41. GPNMB is upregulated in monocyte-derived dendritic cells by BCR-ABL tyrosine kinase inhibitors. PMID: 21874302
  42. Increased GPNMB levels in kidney disease were confirmed by real-time PCR following 5/6 nephrectomy, in streptozotocin-induced diabetes, and in patients with chronic kidney disease. PMID: 21389974
  43. Toxin-conjugated DC-HIL aims to abrogate the ability of Sezary syndrome cells to proliferate in vitro. PMID: 21252093
  44. ADAM10 acts as a sheddase capable of releasing the GPNMB/OA ectodomain from the surface of breast cancer cells. PMID: 20711474
  45. GPNMB expression plays a role in uveal melanoma. PMID: 20375921
  46. GPNMB expression is associated with the basal/triple-negative subtype and is a prognostic marker of poor outcome in patients with breast cancer. PMID: 20215530
  47. GPNMB is a melanosomal protein that is released by proteolytic ectodomain shedding and could serve as a useful and specific histological marker of melanocytic cells. PMID: 20056711
  48. Human OA utilizes the same transcriptional initiation site in both bone and kidney as reported for melanoma cells and is expressed in osteoblast cultures at all stages of differentiation. PMID: 14696968
  49. Cloning and analysis of two fragments in the 5' flanking region of HGFIN; studies indicate p53 cooperates with cytokine-mediated transcription factors to regulate the expression of HGFIN. PMID: 15684612
  50. Osteoactivin is highly expressed in normal and inflammatory liver macrophages, suggesting a significant role in acute liver injury. PMID: 15763343

Show More

Hide All

Database Links

HGNC: 4462

OMIM: 604368

KEGG: hsa:10457

STRING: 9606.ENSP00000371420

UniGene: Hs.190495

Involvement In Disease
Glioblastoma multiforme patients that exhibit increased mRNA and protein levels (>3-fold over normal brain) in biopsy samples have a significantly higher risk of death (PubMed:16609006). In the context of cancer immunotherapy, including that of melanoma, has been proposed to be used as a cell surface protein antigen targeted by antibodies coupled to cytotoxic drugs (PubMed:16489096).
Protein Families
PMEL/NMB family
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Melanosome membrane; Single-pass type I membrane protein. Early endosome membrane; Single-pass type I membrane protein.
Tissue Specificity
Widely expressed, but very low expression, if any, in the brain. Expressed in the epidermis with higher levels in melanocytes compared with keratinocytes and Langerhans cells (at protein level). Expressed in peripheral blood, but not bone marrow mononucle

Q&A

What are the validated applications for GPNMB recombinant monoclonal antibodies?

GPNMB recombinant monoclonal antibodies have been validated for multiple research applications:

ApplicationValidated Cell/Tissue TypesExpected Results
Western BlotU-118-MG and T98G glioblastoma cell linesBands at 76, 95, and 120 kDa
ImmunohistochemistryHuman liver, brain tissuesMembranous and cytoplasmic staining
Flow CytometryCancer cell linesSurface expression quantification
ImmunocytochemistryU-87 MG glioblastoma cellsMembranous and cytoplasmic localization

For Western blot applications, specific bands for GPNMB are typically detected at approximately 95 and 120 kDa under reducing conditions . In immunohistochemistry of human liver, GPNMB staining is specifically localized to Kupffer cells .

How is GPNMB expression regulated in normal versus pathological conditions?

In neurodegenerative contexts:

  • GPNMB protein and mRNA increase as a result of insufficient progranulin in peripheral immune cells at very early ages

  • CSF from GRN-FTD patients shows increased amounts of GPNMB relative to non-demented controls

  • An age-dependent increase in GPNMB expression occurs in mouse models of progranulin deficiency. While 3-month-old Grn KO mice show no significant change, 12-month-old Grn KO mice demonstrate markedly increased GPNMB expression

In oncological contexts:

  • 70% of glioblastoma multiforme (GBM) patient samples are positive for GPNMB transcripts

  • GPNMB expression confers a more migratory and invasive phenotype on breast cancer cells

This differential regulation makes GPNMB an attractive biomarker for disease progression and potential therapeutic intervention.

How do different epitope-targeting GPNMB antibodies compare in experimental applications?

Different antibodies targeting various GPNMB epitopes demonstrate variable experimental utility:

AntibodyTarget EpitopeBinding AffinityOptimal Applications
VH87Recombinant human GPNMBEC₅₀: 6.1 ± 0.09 nM, KD: 13.4 nMCell-based assays
CR011Extracellular domainNot specifiedAntibody-drug conjugates
G11 & U2 (IgG2b mAbs)Varied epitopes0.27-9.6 × 10⁸ M⁻¹Detection applications

The CR011 monoclonal antibody alone did not inhibit melanoma cell growth, but when linked to monomethylauristatin E (MMAE) to create CR011-vcMMAE (glembatumumab vedotin), it potently inhibited GPNMB-positive melanoma cell growth in vitro . When selecting antibodies for specific applications, researchers should consider the accessibility of epitopes under experimental conditions (native vs. denatured protein) and whether post-translational modifications might affect antibody recognition.

What are the critical considerations for validating GPNMB antibody specificity?

Robust validation of GPNMB antibodies requires multiple complementary approaches:

  • Genetic knockout/knockdown validation: Western blot analysis comparing U-87 MG parental and GPNMB-knockdown cell lines should show significantly reduced or absent bands at 76 and 120 kDa in knockdown cells

  • Multiple technique validation: Combining Western blot with immunocytochemistry provides stronger evidence of specificity. In immunocytochemistry experiments, U-87 MG control and GPNMB-knockdown cells labeled with different fluorescent dyes show differential staining patterns with anti-GPNMB antibodies (0.2 μg/mL concentration)

  • Multiple cell line testing: Validation across different GPNMB-expressing cell lines like U-118-MG, T98G, and U-87 MG confirms consistent detection patterns

  • Signal-to-noise analysis: Background staining should be minimal in negative controls, with clear specific staining in positive samples

These validation steps are essential before proceeding to experimental applications, particularly for advanced studies examining GPNMB as a therapeutic target.

How does GPNMB expression correlate with disease progression and patient outcomes?

GPNMB expression demonstrates significant correlations with disease progression in both neurodegenerative and oncological contexts:

In neurodegenerative diseases:

  • Age-dependent increases in GPNMB expression in progranulin-deficient models suggest progressive compensatory mechanisms related to endo-lysosomal dysfunction

  • GPNMB upregulation appears to be an early event in peripheral immune cells that precedes central nervous system pathology

In cancer:

  • Univariate and multivariate analyses show that GBM patients with relatively high mRNA GPNMB transcript levels (>3-fold over normal brain) have a significantly higher risk of death (hazard ratios: 3.0, 2.2, and 2.8)

  • Epithelial-specific GPNMB staining serves as an independent prognostic indicator for breast cancer recurrence

  • GPNMB expression is associated with the basal/triple-negative breast cancer subtype, which typically has poor outcomes

These correlations position GPNMB as both a prognostic biomarker and potential therapeutic target, particularly for aggressive cancer types lacking targeted therapies.

What are the optimal protocols for GPNMB detection in different experimental systems?

Optimal GPNMB detection protocols vary by application:

For Western Blot:

  • Lyse cells in appropriate buffer

  • Separate proteins on SDS-PAGE

  • Transfer to PVDF membrane

  • Block and probe with 0.5 μg/mL anti-GPNMB antibody

  • Detect with HRP-conjugated secondary antibody

  • Expect bands at approximately 76, 95, and 120 kDa

For Immunohistochemistry:

  • Fix tissues in formalin and embed in paraffin

  • Section tissues (typically 4-5 μm)

  • Perform antigen retrieval

  • Apply anti-GPNMB antibody at 3 μg/mL overnight at 4°C

  • Use HRP-DAB detection system and hematoxylin counterstain

For Immunocytochemistry:

  • Fix cells appropriately (e.g., 4% paraformaldehyde)

  • Permeabilize if detecting intracellular epitopes

  • Block non-specific binding

  • Incubate with anti-GPNMB antibody (0.2 μg/mL)

  • Apply fluorescently-labeled secondary antibody

  • Counterstain nuclei with DAPI

Optimization of these protocols for specific experimental contexts is essential, particularly when examining novel tissue types or cell lines.

How should researchers address GPNMB glycosylation patterns in experimental design?

GPNMB is a heavily glycosylated protein, requiring specific methodological considerations:

  • Multiple molecular weight forms: Western blots typically detect GPNMB at 76, 95, and 120 kDa, reflecting different glycosylation states . Researchers should anticipate multiple bands rather than a single discrete band.

  • Deglycosylation controls: Including enzyme-treated samples (PNGase F or similar) can help identify the core protein versus glycosylated forms.

  • Antibody selection: Choose antibodies that recognize epitopes minimally affected by glycosylation patterns. Some antibodies may preferentially detect specific glycoforms.

  • Cell type considerations: Different cell types may produce GPNMB with varying glycosylation patterns. For example, cancer cells often exhibit altered glycosylation compared to normal cells.

  • Functional implications: When studying GPNMB function, consider how glycosylation may affect interactions with binding partners or receptor activation.

Understanding these patterns is particularly important when developing therapeutic approaches targeting GPNMB, as glycosylation may affect antibody accessibility to epitopes.

What are the technical challenges in developing GPNMB-targeted therapeutic antibodies?

Development of GPNMB-targeted therapeutic antibodies faces several technical challenges:

  • Expression heterogeneity: GPNMB expression varies significantly across patients and tissue types. Ectopic overexpression and siRNA studies show that GPNMB expression levels directly correlate with sensitivity to antibody-drug conjugates like CR011-vcMMAE .

  • Dose optimization: The killing effects of dual-targeted bispecific T-cell engagers (DbTEs) on cancer cell lines do not follow standard dose-dependent curves, requiring careful concentration optimization .

  • Glycosylation variability: Multiple glycosylated forms of GPNMB may affect epitope accessibility and antibody binding efficiency.

  • Target validation: In vivo toxicity, specificity, and efficacy studies are essential for accurately assessing therapeutic potential .

  • Combination approaches: GPNMB-targeting antibodies may require combination with other therapeutic modalities to achieve maximal efficacy, particularly in treatment-resistant cancers.

Despite these challenges, GPNMB-targeted therapies show promise. In a melanoma xenograft model, CR011-vcMMAE induced dose-proportional antitumor effects, including complete regressions, at doses as low as 1.25 mg/kg .

How can GPNMB antibodies be applied in investigating neuroinflammatory mechanisms?

GPNMB antibodies offer valuable tools for studying neuroinflammatory mechanisms:

  • Peripheral-central immune crosstalk: GPNMB upregulation in peripheral immune cells of Grn KO mice occurs at early ages, preceding brain pathology, suggesting potential for studying peripheral-central immune communication .

  • Macrophage phenotyping: In Grn KO macrophages, GPNMB upregulation correlates with altered cytokine release profiles. Adding recombinant GPNMB ECD (0.5-1.0 μg/mL) to wild-type peritoneal macrophages mimics the cytokine release phenotype of Grn KO macrophages .

  • Treatment response assessment: GPNMB antibodies can help monitor responses to treatments targeting neuroinflammatory pathways. GPNMB expression may serve as a biomarker for early disease stages before development of CNS pathology .

  • MITF pathway investigation: GPNMB regulation involves the MITF transcription factor, which is dysregulated in Grn KO macrophages. MITF inhibitors like ML329 can be used alongside GPNMB antibodies to explore this regulatory pathway .

These applications highlight the value of GPNMB antibodies beyond cancer research, extending to neurodegenerative disease mechanisms and potential therapeutic strategies.

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.