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:
Cancer types with high ALPP/ALPPL2 expression include:
Ovarian carcinoma
Endometrial carcinoma
Germ cell tumors
Non-small cell lung carcinoma
Gastric carcinoma
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
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.
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:
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.
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:
SDS-PAGE conditions:
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
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
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.
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:
Blocking and antibody incubation:
Block with 10% normal serum (goat or other species matching secondary antibody)
Primary antibody concentrations:
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)
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:
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:
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.
Discrepancies in observed molecular weight for ALPP are common in Western blot experiments and can be systematically addressed:
Common observation:
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:
| Issue | Possible Cause | Solution |
|---|---|---|
| Multiple bands near 70 kDa | Different glycoforms | Treat with PNGase F to remove N-linked glycans |
| Band at unexpected size (much lower) | Proteolytic degradation | Add protease inhibitors freshly to lysis buffer |
| No band visible at either 58 or 70 kDa | Low expression level | Increase protein loading or use immunoprecipitation to enrich |
| Smeared band | Overloaded protein or incomplete denaturation | Reduce 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
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:
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
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
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.
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 category | Examples | Assessment method |
|---|---|---|
| Target expression | ALPP/ALPPL2 levels | IHC, flow cytometry |
| Immune activation | CD8+ T cell infiltration | IHC, flow cytometry |
| Checkpoint expression | PD-L1, LAG-3, TIM-3 levels | IHC, flow cytometry |
| T cell function | IFN-γ, Granzyme B, Perforin | ELISPOT, intracellular staining |
| Soluble mediators | Cytokines, chemokines | Multiplex 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 .
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.
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.
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:
| Application | Validation approach | Positive controls | Negative controls |
|---|---|---|---|
| Western blot | Multiple cell lines/tissues | Placental tissue, JAR cells | ALPP-negative tissues, knockout cells |
| IHC/IF | Multiple FFPE tissues | Third-trimester placenta | Normal tissues except placenta |
| Flow cytometry | Various fixation/permeabilization methods | ALPP-transfected cells | Isotype controls, unstained cells |
| IP-MS | Protein identification confirmation | ALPP-overexpressing cells | IgG 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.
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