PSMA Antibody

Prostate Specific Cancer Antigen, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Evolution of Anti-PSMA Antibodies

AntibodyEpitope TargetKey Features
7E11-C5.3IntracellularFirst murine mAb; limited therapeutic utility due to cytoplasmic binding
J591ExtracellularHumanized mAb; internalizes upon binding, enabling radionuclide/drug delivery
EP192Extracellular dimerRabbit mAb; detects PSMA in formalin-fixed tissues for diagnostics
CC-1 (10B3)Tumor/vasculatureBispecific IgG targeting PSMA and CD3; enhances T-cell recruitment

Key advancements:

  • Transition from murine to humanized/human antibodies to reduce immunogenicity .

  • Development of dimer-specific antibodies (e.g., EP192) with higher diagnostic accuracy .

  • Engineering bispecific antibodies for dual targeting of tumor cells and vasculature .

Diagnostic Imaging

  • ProstaScint® Scan: Uses 7E11-C5.3 conjugated to indium-111 for detecting metastatic prostate cancer .

  • PSMA PET/CT: Next-gen antibodies (e.g., J591) labeled with gallium-68 or fluorine-18 improve sensitivity for micro-metastases .

Targeted Radiotherapy

  • 177Lu-J591: Delivers β-radiation to PSMA+ cells, showing 3-fold survival improvement in preclinical models .

  • 225Ac-PSMA-617: α-emitter achieving >50% PSA reduction in 64% of mCRPC patients .

Antibody-Drug Conjugates (ADCs)

  • ARX517: Anti-PSMA ADC with microtubule inhibitor payload. In the APEX-01 trial (NCT04662580):

    Dose Cohort (mg/kg)PSA50 Response RateGrade ≥3 TRAEs
    1.4–1.725%5–20%
    2.0–2.8852%10–17%
    Deep PSA90 responses (26%) and tumor shrinkage (56% of evaluable patients) were observed .

Mechanistic Insights from Preclinical Studies

  • Internalization Efficiency:

    • J591 exhibits 22.4% internalization within 24 hours, enabling effective drug delivery .

    • Novel antibodies like 11A show 32.1% internalization at 48 hours, surpassing first-gen agents .

  • Neovasculature Targeting: PSMA antibodies bind tumor-associated blood vessels in lung SCC, renal cell carcinoma, and glioblastoma, enabling broad oncology applications .

Emerging Frontiers

  • Combination Therapies: Pairing PSMA antibodies with PARP inhibitors or immunotherapies to overcome resistance .

  • Non-Prostate Cancers: Early trials targeting PSMA+ vasculature in lung SCC and pancreatic adenocarcinoma .

  • CAR-T Cell Therapy: PSMA-directed CAR-T (e.g., P-PSMA-101) induced complete responses in 30% of refractory prostate cancer patients .

Product Specs

Introduction
Prostate-specific membrane antigen (PSMA) is a protein found on the surface of prostate cancer cells. It is also found on the surface of new blood vessels that grow in tumors. PSMA is a potential target for imaging and treating prostate cancer. This is because it is expressed on the surface of prostate cancer cells at much higher levels than on normal cells. PSMA is a type II membrane protein with folate hydrolase activity. While it is primarily found in prostatic epithelium, PSMA expression can also be found in the small bowel and brain.
Formulation
The antibody is supplied as a 1 mg/ml solution in phosphate-buffered saline (PBS) at pH 7.4, containing 0.02% sodium azide and 10% glycerol.
Storage Procedures
Store at 4°C for up to 1 month. For longer storage, store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The antibody is stable for 12 months when stored at -20°C. It is stable for 1 month when stored at 4°C.
Applications
This PSMA antibody has been validated for use in ELISA, Western blot analysis, immunocytochemistry/immunofluorescence (ICC/IF), and flow cytometry. The recommended dilution range for Western blot analysis is 1:1,000 to 1:2,000. For ICC/IF, the recommended dilution is 1:100. For flow cytometry, the recommended dilution is 1:200. Optimal working dilutions should be determined by titration for each application.
Synonyms
PSMA, Prostate Specific Cancer Antigen, PSM, FGCP, FOLH, GCP2, mGCP, GCPII, NAALAD1, NAALAdase, FOLH1, Glutamate carboxypeptidase 2, Glutamate carboxypeptidase II, Membrane glutamate carboxypeptidase, N-acetylated-alpha-linked acidic dipeptidase I, Pteroylpoly-gamma-glutamate carboxypeptidase, Folylpoly-gamma-glutamate carboxypeptidase, Folate hydrolase 1, Prostate-specific membrane antigen, Cell growth-inhibiting protein 27.
Purification Method
PSMA antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
Pk1H7AT.
Immunogen
Anti-human PSMA mAb is derived from hybridization of mouse FO myeloma cells with spleen cells from BALB/c mice immunized with recombinant human PSMA amino acids 117-351 purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and κ light chain.

Q&A

What is PSMA and why is it an important target for antibody development?

PSMA is a type II transmembrane protein expressed in all forms of prostatic tissue, with increased expression in prostate cancer. It has a unique 3-part structure consisting of a 19-amino-acid internal portion, a 24-amino-acid transmembrane portion, and a 707-amino-acid external portion . The PSMA gene is located on the short arm of chromosome 11 in a region not commonly deleted in prostate cancer . Its importance as a target stems from its consistent expression in prostatic tissues, increased expression in malignant cells (80.2% of cells positive in malignant tissue compared to 69.5% in benign epithelial tissue), and its internalization capabilities that allow antibody-drug conjugates to be delivered intracellularly . Additionally, PSMA expression is inversely related to androgen levels, making it potentially more targetable in androgen-independent disease states .

How do PSMA expression patterns differ across tissue types and disease states?

Studies have consistently demonstrated PSMA expression in all types of prostate tissue with increased expression in cancer tissue . PSMA binding occurs in the epithelial cells of the prostate but not in the basal or stromal cells . There is a progressive increase in PSMA staining from benign epithelial tissue (69.5% of cells positive) to high-grade prostatic intraepithelial neoplasia (77.9% of cells positive) to malignant cells (80.2% of cells positive) .

Beyond prostate tissue, PSMA is expressed in duodenal epithelial (brush border) cells and proximal tubule cells in the kidney . Significantly, PSMA is expressed in the neovasculature associated with various cancers including conventional (clear cell) renal cell, transitional cell of the bladder, testicular-embryonal, neuroendocrine, colon, and breast cancers . Interestingly, this neovasculature binding does not seem to occur in prostate cancer itself .

What are the key functional characteristics of PSMA that make it suitable for targeted antibody approaches?

PSMA possesses several functional characteristics that make it an excellent target for antibody-based approaches:

  • Enzymatic activity: PSMA functions as a glutamate-preferring carboxypeptidase , providing a potential functional target.

  • Internalization capability: PSMA contains an internalization signal that allows it to be internalized from the cell surface into an endosomal compartment . This characteristic is particularly important for therapeutic applications as it enables antibody-drug conjugates to deliver cytotoxic agents directly into target cells.

  • Selective expression: The increased expression in malignant cells provides a differential targeting opportunity .

  • Extracellular domain accessibility: The large extracellular portion (707 amino acids) provides abundant epitopes for antibody targeting .

  • Angiogenic marker: Its expression in tumor-associated neovasculature of non-prostatic cancers makes it a potential target for anti-angiogenic strategies .

What are the major types of anti-PSMA antibodies used in research, and how do their binding properties differ?

Several types of anti-PSMA antibodies have been developed for research and clinical applications:

  • First-generation antibody (7E11): Originally developed with LNCaP prostate cancer cell line, mAb 7E11 was the first anti-PSMA antibody. It recognizes and binds to a PSMA intracellular or cytoplasmic epitope . This antibody forms the basis for the FDA-approved ProstaScint scan.

  • Second-generation antibodies: Newer antibodies target the extracellular portion of PSMA and can be internalized by PSMA-expressing cells . These include:

    • J591, J415, J533, and E99: These antibodies demonstrate high-affinity binding to viable PSMA-expressing cells and are rapidly internalized .

    • J591: The most clinically developed deimmunized IgG monoclonal antibody .

    • scFvD2B: An antibody fragment specific for PSMA that has been tested with various radiolabels for imaging applications .

  • Fully human or humanized antibodies: These have been developed to replace murine antibodies, making them more likely to be diagnostically and therapeutically effective without possible antimouse reactions .

  • Dimer-specific epitope antibodies: Recent anti-PSMA antibodies have identified dimer-specific epitopes on PSMA-expressive tumor cells .

What methodologies are used to evaluate the specificity and immunoreactivity of PSMA antibodies?

Evaluation of PSMA antibodies typically involves multiple methodological approaches:

  • Cell-based assays: Using PSMA-expressing cell lines (naturally expressing like LNCaP or transfected like PC3-PIP and LS174T-PSMA) to test binding specificity, with PSMA-negative cell lines (PC3, LS174T) as controls .

  • Immunohistochemical analysis: Applied to evaluate binding patterns in tissue specimens. This approach has demonstrated the correlation between PSMA expression and severity of cancer .

  • Radiolabeling and immunoreactivity assessment: Antibodies are labeled with radioisotopes (e.g., 123I, 111In) and tested for maintained immunoreactivity post-labeling .

  • In vivo biodistribution studies: Analyzing the tissue distribution of radiolabeled antibodies in animal models bearing PSMA-positive and PSMA-negative tumors to assess specific uptake and clearance profiles .

  • Internalization assays: Evaluating the rate and extent of antibody internalization, which is particularly important for antibodies targeting the extracellular domain of PSMA .

How do radiolabeling methods affect the performance of PSMA antibodies for imaging and therapeutic applications?

Radiolabeling methods significantly impact the performance of PSMA antibodies through several mechanisms:

  • Label selection:

    • Diagnostic isotopes (123I, 111In) versus therapeutic isotopes (177Lu)

    • Half-life considerations relative to antibody pharmacokinetics

    • Emission characteristics (gamma, beta) relevant to application

  • Labeling chemistry:

    • Direct iodination versus conjugation-based approaches

    • Impact on immunoreactivity and binding affinity

    • Stability of the radiolabel in vivo

  • Optimization requirements:

    • Purification methods to remove free radioisotope

    • Quality control procedures to ensure consistent specific activity

    • Preservation of immunoreactivity post-labeling

For example, the scFvD2B antibody fragment has been radiolabeled with different isotopes (131I, 111In, 123I) for imaging applications, with optimization required for each labeling approach to maintain specificity and potency of tumor uptake .

How do PSMA antibodies compare in their effectiveness for targeting different stages of prostate cancer progression?

PSMA antibody effectiveness varies across disease stages in important ways:

  • Primary disease: PSMA expression is increased in primary prostate cancer compared to benign tissue, making antibody targeting potentially effective for primary disease detection .

  • Androgen-independent disease: PSMA expression increases as cells become more androgen independent, potentially making PSMA antibodies more effective in later-stage hormone-refractory disease . Denmeade and colleagues demonstrated that PSMA activity in prostate cancer cell lines increased as cells became more androgen independent .

  • Metastatic disease: PSMA expression independently predicts disease recurrence, with overexpressing tumors showing higher recurrence rates (57% vs 28% for non-overexpressing) and shorter time to recurrence (34.78 vs 43.75 months) . This makes PSMA antibodies potentially valuable for detecting metastatic disease.

  • Performance data: The ProstaScint scan (using 7E11 antibody) has shown greatest accuracy for detecting extraprostatic soft tissue disease and less accuracy for detecting bone metastases or disease limited to the prostate bed .

PSMA ExpressionRecurrence RateMean Time to Recurrence (months)P Value
Non-overexpressing (n=71)28% (20/71)43.750.001
Overexpressing (n=65)57% (37/65)34.780.001

What strategies are being explored to enhance the therapeutic efficacy of PSMA-targeted antibody conjugates?

Several strategies are being explored to enhance therapeutic efficacy:

  • Antibody-radionuclide conjugates:

    • Linking antibodies to therapeutic radionuclides (e.g., 177Lu)

    • Early clinical trials have shown radionuclide-antibody compounds can localize to tumor sites, including bony metastases

    • Preclinical studies with 177Lu-labeled mAbs have demonstrated >3-fold increase in survival in animal models

  • Antibody-toxin conjugates:

    • MLN2704: J591 antibody linked to the maytansinoid DM1

    • A5-PE40 and D7-PE40: Recombinant anti-PSMA immunotoxins

    • New linker technologies being developed to improve selective targeting

  • Immunotherapeutic approaches:

    • Creation of artificial T-cell receptors incorporating PSMA-specific single-chain antibodies

    • Dendritic cells pulsed with PSMA peptides to generate immune responses

    • PSMA-based vaccines using recombinant soluble PSMA protein

  • Combination strategies:

    • Using different combinations of anti-PSMA antibodies or combining with antibodies to other targets (GM2, KSA, Thomsen-Friedenreich antigen)

    • This approach aims to decrease nonspecific binding and enhance targeting specificity

  • Molecular engineering:

    • Development of fully human antibodies to reduce immunogenicity

    • Modification of the antibody structure to optimize pharmacokinetics and tissue penetration

    • Manipulation of PSMA promoter regions to develop antiangiogenic gene therapy constructs

How can PSMA antibodies be utilized for targeting tumor-associated neovasculature in non-prostatic malignancies?

PSMA antibodies represent a unique opportunity for targeting tumor-associated neovasculature in non-prostatic malignancies through several approaches:

  • Diagnostic imaging: PSMA antibodies have demonstrated uptake in various non-prostatic malignancies, including incidental findings like renal cell carcinoma, non-Hodgkin's lymphoma, neurofibromatosis, and meningioma . This suggests potential use for broad cancer imaging applications.

  • Anti-angiogenic therapy: Since PSMA is expressed in the neovasculature of numerous cancers (conventional renal cell, transitional cell of bladder, testicular-embryonal, neuroendocrine, colon, and breast), antibodies could be used to deliver therapeutic agents specifically to tumor blood vessels .

  • Combination therapy: Clinical work has explored combining anti-PSMA mAb with interleukin-2 in phase II trials for renal cell cancer, demonstrating the potential for immunomodulatory approaches .

  • Mechanism exploration: Understanding why PSMA is expressed in tumor-associated neovasculature but not in benign vessels could lead to selective antiangiogenic gene therapy constructs .

  • Tissue-specific targeting: Differential expression patterns across tumor types could be exploited for tumor-specific targeting strategies in difficult-to-treat cancers .

What are the optimal preclinical models for evaluating PSMA antibody specificity and efficacy?

Optimal preclinical models for PSMA antibody evaluation include:

  • Cell line selection:

    • Naturally PSMA-expressing lines: LNCaP cells are the gold standard as they naturally express PSMA

    • Engineered PSMA-expressing lines: PC3-PIP and LS174T-PSMA (transfected with PSMA)

    • Negative control lines: PC3 and LS174T (PSMA-negative)

  • Animal models:

    • Subcutaneous xenograft models using PSMA-positive and PSMA-negative cell lines

    • Orthotopic models that better recapitulate the prostate microenvironment

    • Metastatic models for assessing targeting of disseminated disease

    • Patient-derived xenografts that maintain tumor heterogeneity

  • Evaluation parameters:

    • Biodistribution studies to assess targeting specificity

    • Pharmacokinetic and clearance analysis

    • Imaging potential assessment using various modalities

    • Therapeutic efficacy in tumor growth inhibition studies

  • Translational considerations:

    • Models that recapitulate androgen-dependent and androgen-independent states

    • Assessment of neovasculature targeting in non-prostatic tumor models

    • Toxicity evaluation in relevant models to predict clinical safety

What quality control measures are essential when producing and validating PSMA antibodies for research applications?

Essential quality control measures include:

  • Production standards:

    • GMP-compliant production systems (prokaryotic versus eukaryotic)

    • Consistent purification protocols to ensure batch-to-batch reproducibility

    • Purity assessment using methods like SDS-PAGE, HPLC, or capillary electrophoresis

  • Functional validation:

    • Binding affinity determination (KD values)

    • Epitope specificity confirmation

    • Immunoreactivity assessment post-modification (e.g., after radiolabeling)

    • Internalization capacity evaluation for antibodies targeting extracellular domains

  • Physical characterization:

    • Size and aggregation analysis

    • Stability testing under various storage conditions

    • Formulation optimization for research applications

  • Biological validation:

    • Cross-reactivity testing against non-target tissues

    • Specificity testing using PSMA-positive and PSMA-negative controls

    • In vitro functional assays (e.g., antibody-dependent cellular cytotoxicity for therapeutic applications)

    • In vivo validation in appropriate animal models

How can researchers address the challenges of non-specific binding and optimize target-to-background ratios in PSMA antibody applications?

Researchers can address non-specific binding challenges through several approaches:

  • Antibody engineering strategies:

    • Optimization of binding domains to enhance specificity

    • Fragment generation (e.g., scFv) to improve tissue penetration and clearance properties

    • Humanization or deimmunization to reduce immunogenicity while maintaining specificity

  • Experimental design considerations:

    • Inclusion of appropriate blocking agents to reduce non-specific interactions

    • Optimization of antibody concentration and incubation conditions

    • Use of competitive binding assays to confirm specificity

  • Imaging protocol optimization:

    • Timing optimization: Determining optimal imaging timepoints based on pharmacokinetics

    • Background reduction: Developing protocols to enhance target-to-background ratios

    • Signal enhancement: Application of contrast enhancement techniques or dual-labeled approaches

  • Combination approaches:

    • Using multiple anti-PSMA antibodies targeting different epitopes

    • Combining PSMA antibodies with antibodies to other targets to enhance specificity

    • Development of bispecific antibodies to improve targeting precision

  • Advanced production techniques:

    • Selection of expression systems that yield properly folded and glycosylated antibodies

    • Post-production modification to enhance binding characteristics

    • Quality control procedures to ensure consistent specificity across batches

What novel PSMA antibody formats are being developed to improve tumor penetration and reduce immunogenicity?

Novel PSMA antibody formats under development include:

  • Antibody fragments:

    • scFv fragments like scFvD2B that maintain specificity while improving tumor penetration

    • Fab fragments with optimized pharmacokinetic properties

    • Minibodies and diabodies with intermediate size and clearance characteristics

  • Bispecific antibodies:

    • Formats targeting both PSMA and another tumor-associated antigen

    • Bispecific T-cell engagers (BiTEs) that bring immune effector cells to PSMA-expressing tumors

    • Dual-targeting formats that enhance specificity through avidity effects

  • Fully human antibodies:

    • Development using technologies like XenoMouse™ to eliminate immunogenicity

    • Humanization of existing murine antibodies to reduce anti-mouse reactions while preserving binding characteristics

  • Engineered binding domains:

    • Nanobodies derived from camelid antibodies

    • Designed ankyrin repeat proteins (DARPins)

    • Affibodies and other scaffold proteins with high stability and specificity

  • Optimization strategies:

    • Affinity maturation to enhance binding while maintaining specificity

    • Engineering internalization signals to improve intracellular delivery

    • Modification of glycosylation patterns to optimize immune effector functions

How might advances in understanding PSMA's enzymatic functions influence future antibody targeting strategies?

Advances in understanding PSMA's enzymatic functions could influence targeting strategies in several ways:

  • Functional inhibition approaches:

    • Development of antibodies that specifically inhibit PSMA's glutamate-preferring carboxypeptidase activity

    • Understanding the impact of enzymatic inhibition on tumor growth and survival

    • Designing antibodies that bind to catalytic sites versus non-catalytic epitopes

  • Substrate-based targeting:

    • Using knowledge of PSMA's substrate preferences to design prodrugs activated by PSMA

    • Developing antibodies that recognize PSMA-substrate complexes

    • Creating antibody-substrate conjugates with enhanced targeting specificity

  • Structure-function relationships:

    • Utilizing structural knowledge to identify critical domains for antibody targeting

    • Designing antibodies that induce conformational changes affecting enzymatic function

    • Identifying allosteric sites that could be targeted by novel antibody formats

  • Natural ligand discovery:

    • If natural ligands for PSMA are identified, developing antibodies that mimic or block these interactions

    • Understanding how ligand binding affects PSMA internalization and signaling

    • Designing antibody-ligand fusion proteins for enhanced targeting

  • Microenvironment interactions:

    • Exploring how PSMA enzymatic activity affects the tumor microenvironment

    • Developing antibodies that modulate PSMA's interaction with microenvironmental components

    • Understanding how PSMA enzymatic activity contributes to angiogenesis in non-prostatic tumors

What emerging imaging technologies are being paired with PSMA antibodies to enhance detection sensitivity and specificity?

Emerging imaging technologies being paired with PSMA antibodies include:

  • Advanced nuclear medicine techniques:

    • PET imaging with novel radioisotopes optimized for antibody pharmacokinetics

    • SPECT/CT for improved anatomical localization of antibody uptake

    • Theranostic approaches that combine diagnostic and therapeutic radioisotopes

  • Multimodal imaging approaches:

    • Antibodies labeled with both radioisotopes and fluorescent dyes

    • Combination of nuclear and optical imaging for intraoperative guidance

    • Integration of MRI-detectable labels for multiparametric imaging

  • Pretargeting strategies:

    • Two-step approaches that separate antibody targeting from imaging agent delivery

    • Click chemistry-based methods for in vivo conjugation

    • Clearing agents to improve target-to-background ratios

  • Nanoparticle-based platforms:

    • PSMA antibody-decorated nanoparticles carrying imaging agents

    • Multimodal nanoparticles enabling complementary imaging approaches

    • Stimuli-responsive systems for smart imaging

  • Image analysis innovations:

    • Advanced reconstruction algorithms to enhance detection sensitivity

    • Artificial intelligence and machine learning for improved lesion detection

    • Quantitative imaging biomarkers based on PSMA antibody uptake patterns

Product Science Overview

Introduction

Prostate-specific antigen (PSA) is a protein produced primarily by the prostatic epithelium and the epithelial lining of the periurethral glands. It belongs to the kallikrein family of proteases and is secreted into the seminal fluid, where it plays a crucial role in liquefying the gel formed after ejaculation by digesting the major gel-forming proteins .

Prostate-Specific Antigen (PSA)

PSA is a 33 kDa protein that is strongly expressed in both normal and neoplastic prostatic tissue. Although PSA is considered prostate-specific, low levels of PSA and/or PSA gene expression have been detected in some extraprostatic tissues such as normal breast tissue, breast tumors, breast milk, female serum, endometrium, adrenal neoplasms, and renal cell carcinomas .

Mouse Anti-Human PSA Antibody

The monoclonal mouse anti-human PSA antibody, such as Clone ER-PR8, is intended for use in immunohistochemistry (IHC). This antibody labels prostatic epithelium and is a useful aid for the classification of cancer of prostatic origin. Differential classification is aided by the results from a panel of antibodies .

Immunohistochemistry (IHC) and Clinical Applications

IHC is a technique used to visualize the presence and location of proteins in tissue sections. The monoclonal mouse anti-human PSA antibody is used in IHC to detect PSA in tissue samples. This is particularly useful in diagnosing and classifying prostate cancer. The antibody binds specifically to PSA, allowing pathologists to identify prostatic tissue and differentiate it from other types of tissue .

Importance in Cancer Diagnosis

In prostate cancer, the architecture and polarization of the epithelial cells are deranged, leading to active secretion of PSA into the extracellular space and circulation. This makes PSA a valuable biomarker for prostate cancer diagnosis and monitoring. Elevated levels of PSA in the blood can indicate the presence of prostate cancer, although other conditions such as benign prostatic hyperplasia (BPH) and prostatitis can also cause elevated PSA levels .

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.