ALPP Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery timeframes.
Synonyms
ALPP; PLAP; Alkaline phosphatase, placental type; Alkaline phosphatase Regan isozyme; Placental alkaline phosphatase 1; PLAP-1
Target Names
Uniprot No.

Target Background

Function
Alkaline phosphatase is an enzyme capable of hydrolyzing various phosphate compounds.
Gene References Into Functions
  1. A meta-analysis revealed a strong association between elevated serum ALP levels and reduced overall survival (OS) rates in patients with osteosarcoma. This finding suggests that ALP could serve as a valuable prognostic biomarker in these patients. PMID: 29970708
  2. Studies have detected elevated concentrations of PLAP (placental alkaline phosphatase) in the gingival crevicular fluid of individuals with pre-eclampsia, suggesting a potential role for PLAP in this pregnancy complication. PMID: 26988336
  3. Research has shown that SALL4 (a transcription factor) is a highly sensitive marker for MGCTs (malignant germ cell tumors) in the post-treatment setting, exhibiting stronger diffuse nuclear reactivity than PLAP in most cases. While SALL4 lacks absolute specificity, its sensitivity makes it a valuable tool for diagnosis. PMID: 25906119
  4. Quantum-mechanical computational methods have been employed to investigate the catalytic mechanism of human placental alkaline phosphatase (PLAP). These studies utilized an active-site model based on the enzyme's X-ray crystal structure to gain insights into the enzyme's function. PMID: 25409280
  5. Placental explants, but not their conditioned medium, possess the ability to dephosphorylate IGFBP-1 (insulin-like growth factor binding protein-1) through the action of placental alkaline phosphatase, highlighting the enzyme's role in regulating IGFBP-1 activity. PMID: 24856042
  6. Anti-PLAP antibodies have emerged as potential building blocks for the development of targeted therapies. By coupling these antibodies with cytotoxic drugs, radionuclides, or cytokines, researchers aim to deliver these agents specifically to PLAP-expressing cells, enhancing the therapeutic efficacy and minimizing off-target effects. PMID: 24247025
  7. The ALPP gene, which encodes placental alkaline phosphatase, has been identified as a potential contributor to recurrent spontaneous abortion, suggesting a link between ALPP and reproductive health. PMID: 24296104
  8. Studies have explored the diagnostic utility of alpha5(IV) loss, smoothelin expression, and PLAP expression as markers for gastrointestinal smooth muscle neoplasms. These markers offer valuable insights into the identification and characterization of these tumors. PMID: 24043717
  9. p180, a protein involved in cellular processes, has been shown to be essential for the efficient targeting of placental alkaline phosphatase (ALPP) mRNA to the endoplasmic reticulum (ER), indicating a crucial role in the regulation of ALPP expression. PMID: 24019514
  10. Computational analysis of the electrostatic potentials within the active site of human placental alkaline phosphatase suggests that the positive electrostatic environment contributes to the enzyme's ability to differentiate various substrates. PMID: 21910833
  11. Research has revealed a correlation between high serum alkaline phosphatase levels and MMP-9 (matrix metalloproteinase-9) expression, both of which are associated with metastasis in patients with primary osteosarcoma. This finding highlights the potential clinical significance of these markers in predicting tumor behavior. PMID: 22333159
  12. PLAP exerts a positive influence on DNA replication and acts as a proliferative factor in trophoblastic cells. This observation suggests that PLAP plays a crucial role in the growth and development of the placenta. PMID: 21868091
  13. The presence of undifferentiated gonadal tissue in close proximity to tumors, along with immunostaining patterns (PLAP+, OCT3/4+, and CD117/KIT+), suggests that germ cells within these tumors could be a risk factor for gonadal tumors. PMID: 21692598
  14. Research efforts have focused on understanding the catalytic mechanism of human placental alkaline phosphatase. PMID: 21939286
  15. Studies have identified serum alkaline phosphatase, Gleason score, and the intensity of bone metastasis as significant prognostic factors in prostate cancer, impacting time to progression and overall survival. PMID: 19450995
  16. During adrenocortical development, some fetal zone cells survive due to defective apoptosis and develop into childhood ACT (adrenal cortical tumors), retaining certain embryonal characteristics, such as PLAP expression. This finding suggests a link between PLAP expression and the development of childhood ACT. PMID: 21516013
  17. Serum total calcium, serum inorganic phosphate, and serum alkaline phosphatase have demonstrated inverse relationships with age. These findings suggest that these biochemical markers may be influenced by age-related physiological changes. PMID: 20655896
  18. High serum alkaline phosphatase levels are associated with chronic kidney disease, indicating a potential role for this enzyme as a biomarker for kidney health. PMID: 20299338
  19. Differential expression of Pl(1) and Pl(2) (two isoforms of placental alkaline phosphatase) is likely influenced by linkage disequilibrium with the sequence variation rs2014683G>A in the ALPP gene promoter. This variation has been shown to affect allele-specific binding patterns to placental nuclear proteins, suggesting a role in regulating ALPP isoform expression. PMID: 20663553
  20. Structural studies of placental alkaline phosphatase in complex with various ligands, including pNPP (p-nitrophenyl phosphate) and 5'-AMP (adenosine 5'-monophosphate), have provided insights into the enzyme's substrate binding properties and catalytic mechanism. PMID: 20693656
  21. Exposure of human osteoblasts to low magnitudes of tensile strain has been shown to enhance the expression of alkaline phosphatase. This finding suggests a potential role for mechanical forces in regulating bone formation and remodeling. PMID: 19595020
  22. The PLAP D allele, a genetic variant of the ALPP gene, contains two amino acid substitutions: P209R (692C>G) and E429G (1352 A>G). These substitutions may impact the enzyme's activity or other functional properties. PMID: 11857742
  23. Fluorescence resonance energy transfer (FRET) studies have been utilized to determine the proximity of the protein moiety of a GPI-anchored protein to the membrane surface. These investigations provide valuable information about the spatial organization and membrane interactions of GPI-anchored proteins. PMID: 12081485
  24. The glycosylphosphatidylinositol (GPI) anchor of human placental alkaline phosphatase is modified by a beta-N-acetylglucosaminyl phosphate diester residue. This residue serves as a target for aerolysin, a channel-forming toxin. PMID: 12851398
  25. Human placental alkaline phosphatase has been identified as a receptor for Aeromonas sobria hemolysin, a bacterial toxin. This finding suggests a potential role for PLAP in the pathogenesis of bacterial infections. PMID: 15715171
  26. Structural studies of human placental alkaline phosphatase in complex with functional ligands have yielded valuable insights into the enzyme's structure-function relationships and ligand binding mechanisms. PMID: 15946677
  27. An analysis of birth records from an Indian population in Calcutta revealed a correlation between parity (number of previous pregnancies) and placental weight and birth weight. These observations highlight the influence of maternal reproductive history on placental development and birth outcomes. PMID: 16431676
  28. The crystal structure of strontium-substituted human placental alkaline phosphatase revealed that strontium can replace the calcium ion within the enzyme, leading to modifications in the metal coordination. This finding offers insights into the metal binding properties and structural flexibility of PLAP. PMID: 16815919
  29. Research suggests that the activity of GPI-anchored enzymes can be modulated by features of the membrane microenvironment. This observation implies that the surrounding lipid bilayer can influence the function of these membrane-bound enzymes. PMID: 18416535
  30. Serum bilirubin, alkaline phosphatase, and aspartate aminotransferase have proven to be effective biomarkers for identifying patients with primary biliary cirrhosis who are undergoing UDCA (ursodeoxycholic acid) treatment and are at risk of death or liver transplantation. This finding underscores the importance of these biochemical markers in clinical management. PMID: 18752324
  31. Elevated levels of alkaline phosphatase have been linked to therapy-related pediatric cancer, suggesting a potential role for ALP as a biomarker or indicator of treatment response. PMID: 18802949
Database Links

HGNC: 439

OMIM: 171800

KEGG: hsa:250

STRING: 9606.ENSP00000375881

UniGene: Hs.284255

Protein Families
Alkaline phosphatase family
Subcellular Location
Cell membrane; Lipid-anchor, GPI-anchor.
Tissue Specificity
Detected in placenta (at protein level).

Q&A

What is ALPP and why is it significant in cancer research?

ALPP (alkaline phosphatase placental type) is a cell membrane-attached phosphatase that plays key roles in nucleotide recycling. Its significance in cancer research stems from its highly restricted normal tissue expression pattern combined with high expression in several solid tumor types. ALPP and its related protein ALPPL2 are normally only expressed in placental trophoblasts during fetal development, with minimal expression in other normal tissues, making them ideal candidates for targeted cancer therapies .

Normal tissue expression is primarily limited to:

  • Placental trophoblasts

  • Low levels in reproductive tissue

  • Minimal expression in lung tissue

Cancer types with high ALPP/ALPPL2 expression include:

  • Ovarian carcinoma

  • Endometrial carcinoma

  • Germ cell tumors

  • Non-small cell lung carcinoma

  • Gastric carcinoma

  • Mesothelioma

How do I select the appropriate anti-ALPP antibody for my specific application?

Selection of the appropriate anti-ALPP antibody should be based on your specific experimental needs:

For Western Blot applications:

  • Consider antibodies validated for WB such as Boster's A01718 (for human and mouse samples) or Bio-Techne's MAB5905 (specific for human)

  • Verify the antibody's specificity by checking cross-reactivity data with other alkaline phosphatase family members

  • For human samples, Bio-Techne's MAB5905 shows no cross-reactivity with other related proteins like ALPI or ALPL

For immunohistochemistry/immunofluorescence:

  • Rabbit anti-ALPP antibodies like A01718 have been validated for IHC and IF on placental and tumor tissues

  • Consider antibody clones specifically validated for FFPE tissues if working with archived samples

  • Verify whether antigen retrieval is required (some ALPP antibodies work without special pretreatment)

For flow cytometry:

  • Choose antibodies validated specifically for flow cytometry applications

  • Consider antibodies with recommended dilutions for flow cytometry (e.g., Boster's A01718 has been validated for this application)

The choice between monoclonal and polyclonal antibodies should depend on your specific needs:

  • Monoclonal antibodies offer higher specificity for a single epitope

  • Polyclonal antibodies may provide stronger signal by recognizing multiple epitopes

What is the difference between ALPP and ALPPL2, and do antibodies distinguish between them?

ALPP (placental alkaline phosphatase) and ALPPL2 (placental-like alkaline phosphatase 2) are closely related proteins that share significant homology:

Key differences:

  • ALPP is encoded by the ALPP gene, while ALPPL2 is encoded by a separate gene

  • Both proteins function as alkaline phosphatases and can form homo- and heterodimers

  • They share similar tissue expression patterns, being highly expressed in tumors with minimal normal tissue expression except for placenta

Antibody cross-reactivity:
Most commercially available antibodies fall into two categories:

  • ALPP-specific antibodies: These recognize epitopes unique to ALPP and don't cross-react with ALPPL2

  • ALPP/ALPPL2 dual-reactive antibodies: These recognize epitopes common to both proteins

For example:

  • SGN-ALPV utilizes the humanized antibody h12F3 that is highly specific for both human and cynomolgus monkey ALPP and ALPPL2 proteins but does not recognize other related phosphatases

  • Some antibodies like Bio-Techne's MAB5905 are specifically tested against other alkaline phosphatase family members to verify specificity

When selecting an antibody, verify from the manufacturer whether it distinguishes between ALPP and ALPPL2 or recognizes both, depending on your experimental needs.

How can I accurately quantify ALPP antigen density on the cell surface for ADC development studies?

Accurate quantification of ALPP antigen density is crucial for antibody-drug conjugate (ADC) development and optimization. A methodologically sound approach involves quantitative flow cytometry:

Recommended protocol based on published methods:

  • Direct antibody labeling: Label anti-ALPP antibody and control antibody with a fluorophore (e.g., Alexa Fluor 647)

  • Standard curve generation: Use Simply Cellular anti-Human IgG beads with known antibody binding capacity

  • F/P ratio determination: Calculate the fluorophore/protein ratio of the labeled antibody

  • Cell staining and analysis: Incubate target cells with labeled antibodies and analyze by flow cytometry

  • Conversion to absolute values:

    • Convert Mean Fluorescence Intensity (MFI) to Molecules of Equivalent Soluble Fluorochrome (MESF) using Quantum beads

    • Calculate antibody binding sites per cell by dividing MESF by the F/P ratio

Important considerations:

  • Always include appropriate isotype controls

  • Include both positive control cells (known ALPP expressors like JAR choriocarcinoma cells) and negative control cells

  • For comparative studies across cell lines, normalize for cell size differences

  • When evaluating potential ADC targets, cell surface densities >50,000 copies/cell are generally considered favorable for ADC development

This quantitative approach provides more actionable data than simple "positive/negative" classification and allows for rational selection of optimal target cell populations for therapy development.

What are the optimal conditions for detecting ALPP by Western blot considering its glycosylation status?

ALPP has a theoretical molecular weight of approximately 57-58 kDa, but consistently runs at approximately 70-72 kDa on SDS-PAGE due to post-translational modifications, particularly glycosylation . This discrepancy needs to be considered when optimizing Western blot protocols:

Recommended optimized protocol:

  • Sample preparation:

    • Use RIPA or similar buffer with protease inhibitors

    • For glycosylation studies, consider treating a sample with PNGase F to remove N-linked glycans

    • Include positive control (placental tissue or JAR/JEG-3 cell lysates)

  • SDS-PAGE conditions:

    • Use 8-10% polyacrylamide gels to achieve good separation around 70 kDa

    • Run at 70V (stacking gel)/90V (resolving gel) for 2-3 hours for optimal separation

  • Transfer and detection:

    • Transfer to nitrocellulose membrane at 150 mA for 50-90 minutes

    • Block with 5% non-fat milk/TBS for 1.5 hours at room temperature

    • Incubate with primary anti-ALPP antibody (0.5-2 μg/mL) overnight at 4°C

    • Wash with TBS-0.1% Tween (3× for 5 minutes each)

    • Incubate with HRP-conjugated secondary antibody (1:5000 dilution) for 1.5 hours at room temperature

    • Develop using enhanced chemiluminescence (ECL)

Troubleshooting glycosylation-related issues:

  • If diffuse bands are observed, consider deglycosylation treatments

  • For comparison of protein backbone across samples, parallel runs with and without deglycosylation can be informative

  • Expected band size without glycosylation is closer to the theoretical 58 kDa

  • For JAR or JEG-3 cell lysates, a specific band should be detected at approximately 70 kDa

How do ALPP antibodies perform in detecting circulating tumor cells (CTCs) expressing ALPP/ALPPL2?

Detecting circulating tumor cells (CTCs) using ALPP antibodies represents an advanced application with specific methodological considerations:

Methodological approaches:

  • Flow cytometry-based CTC detection:

    • Direct staining with fluorophore-conjugated anti-ALPP antibodies

    • Multi-parameter flow cytometry combining ALPP with epithelial markers (EpCAM, cytokeratins) and excluding hematopoietic markers (CD45)

    • Expected sensitivity based on available data: 1 CTC per 10^6 peripheral blood mononuclear cells

  • Immunomagnetic separation followed by immunofluorescence:

    • Enrich CTCs using anti-ALPP antibody-coated magnetic beads

    • Confirm CTC identity with immunofluorescence microscopy using separate anti-ALPP antibody clones

    • Include DAPI nuclear staining and exclude CD45+ cells

Technical considerations:

  • Pre-analytical variables (sample collection, processing time) significantly impact CTC recovery

  • Anti-ALPP antibodies should be validated specifically for rare cell detection

  • For live cell isolation, non-toxic antibody clones that maintain target cell viability should be selected

  • Combinations with other tumor-specific markers may increase sensitivity and specificity

The use of ALPP antibodies for CTC detection is particularly promising for tumors known to express ALPP highly, such as germ cell tumors, ovarian and endometrial carcinomas.

What are the optimal conditions for immunohistochemical detection of ALPP in different tissue types?

Optimizing immunohistochemical (IHC) detection of ALPP requires careful consideration of tissue type, fixation, and antibody selection:

General IHC protocol for ALPP detection:

  • Tissue preparation:

    • FFPE (formalin-fixed paraffin-embedded) sections: 4-5 μm thickness

    • Fresh frozen sections: 6-8 μm thickness

  • Antigen retrieval options:

    • For some ALPP antibodies, no special pretreatment is required

    • Heat-mediated antigen retrieval in EDTA buffer (pH 8.0) is effective for many ALPP antibodies

    • For challenging samples, enzymatic retrieval using IHC enzyme antigen retrieval reagent can be considered

  • Blocking and antibody incubation:

    • Block with 10% normal serum (goat or other species matching secondary antibody)

    • Primary antibody concentrations:

      • For paraffin sections: 0.25-0.5 μg/ml or 2-5 μg/ml depending on the antibody

      • Incubate overnight at 4°C or 30 minutes at room temperature

    • Secondary detection:

      • Peroxidase-conjugated or fluorophore-conjugated secondary antibodies

      • For brightfield microscopy: develop with DAB

      • For fluorescence: use appropriate fluorophore-conjugated secondary antibodies

Tissue-specific considerations:

  • Placenta (positive control): Syncytiotrophoblasts show strong membrane staining

  • Tumor tissues: Expression patterns may vary; membranous and/or cytoplasmic staining

  • Normal tissues: Expect minimal staining except in placenta and possibly low levels in lung

  • Mesothelioma: Both epithelioid and sarcomatoid subtypes show ALPP/ALPPL2 expression

Controls to include:

  • Positive control: Placental tissue (third trimester)

  • Negative controls:

    • Primary antibody omission

    • Isotype control antibody

    • Normal tissues (except placenta)

How do I optimize ALPP antibodies for flow cytometry applications in detecting tumor cells?

Optimizing flow cytometry with ALPP antibodies for tumor cell detection requires careful attention to antibody selection, staining protocols, and controls:

Recommended optimization strategy:

  • Antibody selection and preparation:

    • Choose antibodies validated for flow cytometry (e.g., Boster's A01718)

    • For direct detection, consider custom-conjugating purified antibodies with fluorophores suitable for your cytometer configuration

    • For indirect detection, select secondary antibodies with minimal spectral overlap with other fluorophores in your panel

  • Cell preparation protocol:

    • Live cells: Use gentle fixation (1-2% PFA) or stain unfixed cells if antibody recognizes extracellular epitope

    • Fixed cells: 4% paraformaldehyde fixation followed by permeabilization if needed

    • Critical cell concentration: 1×10^6 cells per 100 μL staining reaction

  • Staining protocol optimization:

    • Titrate antibody concentrations (typical range: 0.1-10 μg/mL)

    • Optimize incubation conditions (temperature: 4°C vs. room temperature; time: 15-60 minutes)

    • Include blocking step with 10% normal serum to reduce non-specific binding

    • For multi-color panels, include fluorescence-minus-one (FMO) controls

  • Data acquisition considerations:

    • Adjust voltage settings using unstained and single-stained controls

    • Collect sufficient events (≥10,000 for abundant populations; ≥100,000 for rare populations)

    • Include viability dye to exclude dead cells

Example optimized protocol based on published methods:

  • Fix cells with 4% paraformaldehyde

  • Block with 10% normal goat serum

  • Incubate with rabbit anti-ALPP antibody (1 μg per 1×10^6 cells) for 30 min at 20°C

  • Wash cells 3× with flow buffer (PBS + 2% FBS)

  • Incubate with fluorophore-conjugated secondary antibody (e.g., DyLight 488-conjugated goat anti-rabbit IgG) at appropriate dilution

  • Include proper controls:

    • Isotype control antibody (rabbit IgG) at same concentration

    • Unstained sample

What are the key considerations for developing ALPP/ALPPL2-targeted antibody-drug conjugates (ADCs)?

Developing effective antibody-drug conjugates (ADCs) targeting ALPP/ALPPL2 requires careful consideration of multiple factors:

1. Antibody selection criteria:

  • Binding affinity (sub-nanomolar KD preferred)

  • Specificity for ALPP/ALPPL2 without cross-reactivity to other phosphatases

  • Efficient internalization upon target binding

  • Stability in circulation

  • Low immunogenicity potential

2. Linker-payload selection:

  • Cleavable linkers: Such as the protease-cleavable peptide linker used in SGN-ALPV that allows for release of the cytotoxic payload in the lysosomal environment

  • Payload options:

    • Microtubule inhibitors (e.g., MMAE as used in SGN-ALPV)

    • DNA-damaging agents

    • RNA polymerase inhibitors

  • Drug-to-antibody ratio (DAR): Optimize for balance between potency and pharmacokinetic properties

3. Evaluating ADC efficacy:

  • In vitro assays:

    • Cytotoxicity against ALPP/ALPPL2-positive vs. negative cell lines

    • Internalization rate assessment

    • Mechanism of action studies (cell cycle arrest, apoptosis induction)

    • Immunogenic cell death markers assessment

  • In vivo models:

    • Cell line-derived xenografts

    • Patient-derived xenografts

    • Dose escalation studies

    • Schedule optimization

    • Combination therapies

Example ADC development approach:
SGN-ALPV has demonstrated significant potential as an investigational vedotin ADC:

  • Uses humanized IgG1 monoclonal antibody (h12F3) with high specificity for ALPP/ALPPL2

  • Conjugated to monomethyl auristatin E (MMAE) via a protease-cleavable peptide linker

  • Upon binding, ALPP/ALPPL2 are internalized to lysosomal vesicles

  • Released MMAE drives mitotic arrest, apoptosis, and immunogenic cell death

  • Additional mechanisms include antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP)

This approach has shown robust antitumor activity in preclinical studies against different tumor types expressing ALPP/ALPPL2.

How do I resolve discrepancies in ALPP molecular weight observed in Western blot experiments?

Discrepancies in observed molecular weight for ALPP are common in Western blot experiments and can be systematically addressed:

Common observation:

  • Theoretical molecular weight: 57-58 kDa

  • Observed molecular weight on SDS-PAGE: 70-72 kDa

Key factors contributing to molecular weight discrepancies:

  • Post-translational modifications:

    • Glycosylation is the primary contributor to the higher observed molecular weight

    • ALPP contains multiple glycosylation sites, adding approximately 12-15 kDa

  • Sample preparation effects:

    • Denaturation conditions (reducing vs. non-reducing)

    • Heat treatment duration and temperature

  • Gel system variations:

    • Percentage of acrylamide

    • Buffer systems (Tris-glycine vs. Tris-tricine)

    • Commercial pre-cast vs. laboratory-prepared gels

Systematic troubleshooting approach:

IssuePossible CauseSolution
Multiple bands near 70 kDaDifferent glycoformsTreat with PNGase F to remove N-linked glycans
Band at unexpected size (much lower)Proteolytic degradationAdd protease inhibitors freshly to lysis buffer
No band visible at either 58 or 70 kDaLow expression levelIncrease protein loading or use immunoprecipitation to enrich
Smeared bandOverloaded protein or incomplete denaturationReduce protein amount or increase SDS and heating time

Verification strategies:

  • Run parallel samples with and without deglycosylation treatment

  • Include known positive controls (placental tissue lysate or JAR/JEG-3 cell lysates)

  • Confirm identity by immunoprecipitation followed by mass spectrometry

  • Validate with multiple antibodies recognizing different epitopes

What are the potential causes of cross-reactivity when using ALPP antibodies, and how can they be mitigated?

Cross-reactivity can significantly impact experimental results when working with ALPP antibodies. Understanding and mitigating these issues is essential:

Common sources of cross-reactivity:

  • Related alkaline phosphatase family members:

    • ALPL (tissue non-specific alkaline phosphatase)

    • ALPI (intestinal alkaline phosphatase)

    • ALPPL2 (placental-like alkaline phosphatase 2)

    • These share 87-98% sequence homology in certain regions

  • Species cross-reactivity:

    • Human vs. mouse vs. primate ALPP proteins

    • While some antibodies are species-specific, others may cross-react

  • Non-specific binding:

    • Fc receptor binding in immune cells

    • Endogenous biotin (when using biotin-streptavidin detection systems)

    • Endogenous peroxidases (in IHC/ICC with HRP detection)

Mitigation strategies:

Cross-reactivity sourceVerification methodMitigation strategy
Related phosphatasesTest on recombinant proteins of each family memberChoose antibodies specifically tested against related proteins (e.g., Bio-Techne's MAB5905)
Species cross-reactivityTest on tissues from different speciesUse species-specific antibodies when working with mixed species samples
Fc receptor bindingCompare F(ab')2 vs. whole IgG binding patternsUse F(ab')2 fragments or add Fc block
Endogenous peroxidasesRun control without primary antibodyQuench endogenous peroxidases (3% H2O2 treatment)
Endogenous biotinTest biotin blocking kit effectivenessUse biotin blocking kit or alternative detection system

Practical approach to verify antibody specificity:

  • Include knockout/knockdown controls when possible

  • Perform peptide competition assays with the immunizing peptide

  • Compare staining patterns with multiple antibodies targeting different epitopes

  • Include appropriate isotype controls at matching concentrations

How can I differentiate between ALPP and ALPPL2 expression in tumor samples?

Differentiating between ALPP and ALPPL2 expression in tumor samples requires strategic approaches due to their high homology:

Methodological approaches:

  • Antibody-based differentiation:

    • Use antibodies specifically validated to distinguish between ALPP and ALPPL2

    • Perform sequential staining with different antibodies on serial sections

    • Consider dual immunofluorescence with antibodies against unique epitopes

  • Nucleic acid-based differentiation:

    • Design PCR primers targeting unique regions of each gene

    • Perform quantitative RT-PCR with gene-specific primers

    • RNA in situ hybridization with probes specific to unique regions

    • RNA-seq analysis focusing on distinguishing SNPs or unique exons

  • Protein-based differentiation:

    • Mass spectrometry-based proteomics targeting peptides unique to each protein

    • Isoelectric focusing followed by Western blot (the proteins have slightly different pI values)

Decision tree for differentiation strategy:

  • If working with fixed tissues:

    • Try RNAscope in situ hybridization with gene-specific probes

    • If unavailable, use antibodies reported to distinguish the proteins

  • If working with frozen tissues or cells:

    • Extract RNA for RT-qPCR with gene-specific primers

    • Extract protein for Western blot with discriminating antibodies

    • Consider immunoprecipitation followed by mass spectrometry

  • If working with live cells:

    • Flow cytometry with carefully validated antibodies

    • Single-cell RT-PCR for definitive molecular identification

Important considerations:

  • In many tumor contexts, both proteins may be co-expressed

  • Their functional roles appear similar, so distinguishing them may be more relevant for basic research than therapeutic targeting

  • For therapeutic applications like ADCs, dual-reactive antibodies like h12F3 (used in SGN-ALPV) that recognize both proteins may actually be advantageous

How are ALPP/ALPPL2 antibodies being utilized in emerging cancer immunotherapy approaches?

ALPP and ALPPL2 antibodies are being leveraged in several innovative immunotherapeutic approaches:

1. Chimeric Antigen Receptor (CAR) T-cell therapy:

  • Second-generation CAR T-cells with fully human single-chain variable fragments (scFvs) against ALPP have demonstrated efficient killing of ALPP-expressing tumor cells in preclinical models

  • ALPP-CAR-T cells have shown potent cytotoxicity toward cancer cells

  • Combination strategies pairing ALPP-CAR-T cells with checkpoint inhibitors (anti-PD-1, PD-L1, or LAG-3) have demonstrated enhanced therapeutic efficacy

  • A clinical trial using α-ALPP CAR T cells for ovarian and endometrial cancer has been initiated

2. Antibody-Drug Conjugates (ADCs):

  • SGN-ALPV, a novel investigational vedotin ADC targeting ALPP/ALPPL2, is being evaluated for various solid tumors

  • The ADC consists of a humanized IgG1 monoclonal antibody conjugated to monomethyl auristatin E (MMAE) via a protease-cleavable linker

  • Preclinical studies show robust antitumor activity in cell line and patient-derived xenograft models

  • A clinical study evaluating ALPP/ALPPL2 antibody-drug conjugate in advanced solid tumors is ongoing

3. Bi-specific T-cell Engagers (BiTEs):

  • Emerging research is exploring ALPP-targeted BiTEs that simultaneously bind ALPP on tumor cells and CD3 on T cells

  • This approach aims to recruit and activate T cells at tumor sites without requiring ex vivo manipulation

4. Antibody-based Imaging:

  • Radiolabeled ALPP antibodies are being developed for tumor imaging

  • These could enable non-invasive assessment of ALPP expression to guide patient selection for ALPP-targeted therapies

The high tumor specificity of ALPP/ALPPL2 expression makes these proteins particularly attractive targets for these emerging immunotherapeutic approaches, with minimal risk of on-target/off-tumor toxicity.

What are the key methodological considerations for evaluating ALPP antibodies in combination with immune checkpoint inhibitors?

Evaluating ALPP antibodies in combination with immune checkpoint inhibitors requires careful experimental design:

In vitro assessment methodologies:

  • Co-culture systems:

    • Establish co-cultures of ALPP-expressing tumor cells with immune cells (PBMCs or isolated T cells)

    • Add ALPP antibodies alone or in combination with checkpoint inhibitors

    • Measure:

      • T cell activation markers (CD69, CD25, CD137)

      • Cytokine production (IFN-γ, TNF-α, IL-2)

      • Tumor cell killing (cytotoxicity assays)

      • Immune cell proliferation

  • 3D spheroid/organoid models:

    • Generate 3D cultures of ALPP-positive tumor cells

    • Add immune components and test antibody combinations

    • Assess infiltration and activation of immune cells within 3D structures

In vivo experimental design:

  • Mouse model selection:

    • Syngeneic models engineered to express human ALPP

    • Humanized mouse models with human immune system components

    • Patient-derived xenograft models in immunocompromised mice reconstituted with human immune cells

  • Treatment schedule optimization:

    • Sequential vs. concurrent administration

    • Dose-ranging studies for both agents

    • Duration of treatment and monitoring period

  • Comprehensive endpoint analysis:

    • Tumor growth/regression measurements

    • Immune cell infiltration (flow cytometry, IHC)

    • Cytokine profiles in tumor microenvironment

    • Toxicity assessments

    • Secondary tumor challenge to assess memory response

Biomarker assessment:

Biomarker categoryExamplesAssessment method
Target expressionALPP/ALPPL2 levelsIHC, flow cytometry
Immune activationCD8+ T cell infiltrationIHC, flow cytometry
Checkpoint expressionPD-L1, LAG-3, TIM-3 levelsIHC, flow cytometry
T cell functionIFN-γ, Granzyme B, PerforinELISPOT, intracellular staining
Soluble mediatorsCytokines, chemokinesMultiplex assays, ELISA

The combination of ALPP-CAR-T cells with checkpoint inhibitors targeting PD-1, PD-L1, or LAG-3 has shown increased therapeutic efficacy in preclinical models, warranting further investigation of these combinatorial approaches .

What novel methodologies are being developed for detecting circulating ALPP/ALPPL2 as cancer biomarkers?

Several innovative methodologies are being developed to detect circulating ALPP/ALPPL2 as cancer biomarkers:

1. Liquid biopsy approaches:

  • Circulating tumor cells (CTCs):

    • Microfluidic chip-based enrichment followed by ALPP antibody detection

    • Automated rare cell detection systems combining ALPP with other markers

    • Single-cell molecular profiling of ALPP-positive CTCs

  • Circulating tumor DNA (ctDNA):

    • Detection of ALPP/ALPPL2 gene amplifications in cell-free DNA

    • Methylation analysis of ALPP/ALPPL2 promoter regions in ctDNA

    • Integration with other genomic alterations common in ALPP-expressing tumors

  • Extracellular vesicles (EVs):

    • Isolation of tumor-derived EVs using ALPP antibodies

    • Multiplex analysis of EV cargo from ALPP-positive vesicles

    • EV-based functional assays to assess tumor-immune interactions

2. Advanced detection technologies:

  • Digital ELISA platforms:

    • Single molecule array (Simoa) technology for ultra-sensitive detection of soluble ALPP

    • Multiplex digital protein assays combining ALPP with other tumor markers

  • Mass cytometry (CyTOF):

    • Metal-tagged ALPP antibodies for high-dimensional analysis

    • Integration with other cellular markers for comprehensive phenotyping

  • Aptamer-based detection:

    • Development of ALPP-specific aptamers with potentially superior tissue penetration

    • Combination of aptamers with nanoparticle-based detection systems

3. Point-of-care testing development:

  • Lateral flow immunoassays:

    • Rapid test formats using highly specific ALPP antibodies

    • Enhanced sensitivity through signal amplification technologies

  • Electrochemical biosensors:

    • Antibody-functionalized electrodes for ALPP detection

    • Integration with smartphone-based readers for accessible testing

Methodological validation considerations:

  • Comparison with established tissue-based detection methods

  • Concordance analysis between circulating markers and tumor expression

  • Longitudinal assessment to determine utility for treatment monitoring

  • Correlation with clinical outcomes to establish prognostic/predictive value

These emerging methodologies could potentially transform ALPP/ALPPL2 from tissue-based biomarkers to circulating biomarkers accessible through minimally invasive procedures.

What are the current limitations of ALPP antibodies in research and clinical applications?

Despite their significant potential, ALPP antibodies face several important limitations:

Technical limitations:

  • Variable specificity between ALPP and closely related ALPPL2

  • Limited standardization across research laboratories using different antibody clones

  • Potential cross-reactivity with other alkaline phosphatase family members

  • Challenges in detecting low expression levels in certain tumor types

  • Variability in glycosylation patterns affecting antibody binding efficiency

Research application limitations:

  • Incomplete characterization of expression across tumor types and subtypes

  • Limited understanding of the functional significance of ALPP/ALPPL2 in tumor biology

  • Need for better predictive models to identify responders to ALPP-targeted therapies

  • Challenges in developing animal models that accurately recapitulate human ALPP expression patterns

Clinical development challenges:

  • Limited clinical-grade antibody options validated for companion diagnostic use

  • Need for standardized scoring systems for ALPP/ALPPL2 positivity in patient samples

  • Potential for acquired resistance mechanisms to ALPP-targeted therapies

  • Requirements for extensive safety monitoring due to potential expression in reproductive tissues

Future research priorities to address limitations:

  • Development of antibodies with improved specificity and sensitivity

  • Standardization of detection protocols across laboratories

  • Expanded profiling of ALPP/ALPPL2 expression across tumor types and normal tissues

  • Deeper understanding of ALPP/ALPPL2 biology and function in cancer

  • Development of companion diagnostics alongside therapeutic applications

These limitations represent important areas for future research and development to fully realize the potential of ALPP antibodies in both research and clinical applications.

How can researchers validate new ALPP antibodies for specificity and performance across various applications?

A comprehensive validation strategy for new ALPP antibodies should include:

1. Epitope and specificity characterization:

  • Epitope mapping to determine the recognized region

  • Cross-reactivity testing against:

    • Related proteins (ALPPL2, ALPI, ALPL)

    • Species orthologs (human, mouse, primate)

  • Peptide competition assays to confirm specificity

  • Testing on ALPP/ALPPL2 knockout or knockdown models

2. Application-specific validation:

ApplicationValidation approachPositive controlsNegative controls
Western blotMultiple cell lines/tissuesPlacental tissue, JAR cellsALPP-negative tissues, knockout cells
IHC/IFMultiple FFPE tissuesThird-trimester placentaNormal tissues except placenta
Flow cytometryVarious fixation/permeabilization methodsALPP-transfected cellsIsotype controls, unstained cells
IP-MSProtein identification confirmationALPP-overexpressing cellsIgG control IP

3. Quantitative performance metrics:

  • Sensitivity: Limit of detection in each application

  • Dynamic range: Linear range of signal intensity

  • Reproducibility: Intra- and inter-assay coefficient of variation

  • Lot-to-lot consistency: Testing multiple antibody lots

4. Comparative benchmarking:

  • Side-by-side comparison with established antibody clones

  • Testing across multiple experimental conditions

  • Performance in multiplexed applications

5. Functional validation:

  • Antibody effects on ALPP enzymatic activity

  • Internalization assays for ADC development

  • Effects on cell proliferation/viability (if any)

Documentation and reporting standards:

  • Detailed protocols for each successful application

  • Complete description of validation experiments and results

  • Raw data availability for independent assessment

  • Registration in antibody validation repositories

This multi-faceted validation approach ensures that new ALPP antibodies meet the rigorous standards required for research applications and potential clinical development.

What future research directions hold the most promise for ALPP/ALPPL2-targeted therapies?

Several promising research directions for ALPP/ALPPL2-targeted therapies warrant further investigation:

1. Next-generation antibody-drug conjugates:

  • Novel payload classes beyond microtubule inhibitors

  • Site-specific conjugation technologies for improved homogeneity

  • Dual-targeting ADCs recognizing ALPP/ALPPL2 and a second tumor antigen

  • Development of ADCs with improved therapeutic window

2. Advanced cellular therapies:

  • Allogeneic ALPP-CAR T cell approaches

  • ALPP-CAR NK cells with enhanced persistence

  • CAR-macrophage therapies targeting phagocytosis of tumor cells

  • T cell receptor (TCR)-engineered T cells targeting ALPP/ALPPL2-derived peptides

  • Logic-gated CAR designs requiring dual antigen recognition

3. Combination therapeutic strategies:

  • ALPP-targeted therapies with DNA damage response inhibitors

  • Combinations with emerging immune checkpoint inhibitors (beyond PD-1/PD-L1)

  • Integration with conventional therapies (chemotherapy, radiation)

  • Sequencing strategies for optimal clinical benefit

4. Novel modalities:

  • Proteolysis-targeting chimeras (PROTACs) targeting ALPP/ALPPL2

  • mRNA-based therapeutic approaches

  • Oncolytic viruses engineered to selectively replicate in ALPP-expressing cells

  • Radioimmunoconjugates for theranostic applications

5. Personalized medicine approaches:

  • Development of companion diagnostics for patient selection

  • Identification of biomarkers predictive of response

  • Real-time monitoring of treatment efficacy

  • Adaptive trial designs based on early biological response

Key upcoming milestones:

  • Results from ongoing clinical trials of SGN-ALPV antibody-drug conjugate

  • Data from the α-ALPP CAR T cell trial in ovarian and endometrial cancer

  • Expanded tumor profiling to identify additional cancer types with ALPP/ALPPL2 expression

  • Development of standardized detection methods for patient selection

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.