ROR1 Recombinant Monoclonal Antibody is a laboratory-engineered immunoglobulin designed to bind with high specificity to the extracellular domain of ROR1. Its recombinant nature ensures batch-to-batch consistency, while monoclonal properties guarantee epitope uniformity. ROR1’s role in cancer progression—via Wnt signaling, EMT regulation, and drug resistance—makes this antibody a strategic candidate for targeted therapies .
The synthesis involves:
Immunogen Preparation: Recombinant human ROR1 protein is used to immunize animals .
B Cell Isolation: Splenocytes from immunized hosts are fused with myeloma cells to generate hybridomas .
Cloning and Expression: Antibody genes are cloned into vectors (e.g., pETDuet) and expressed in prokaryotic or mammalian systems .
Purification: Affinity chromatography yields >95% pure IgG1 or Fab fragments .
Ovarian Cancer: ROR1-cFab (chimeric Fab) reduced proliferation by 40% and migration by 60% in A2780 cells via apoptosis induction .
Hepatocellular Carcinoma (HCC): Antibodies like 4A5 inhibited EMT, decreased proliferation (p < 0.01), and reversed chemoresistance by downregulating MDR1 .
Chronic Lymphocytic Leukemia (CLL): Anti-ROR1 mAbs (e.g., D10) blocked leukemic cell engraftment in xenografts by 80% .
ROR1-targeted antibody-drug conjugates (ADCs) and immunotherapies are under evaluation:
Therapy | Type | Target Indications | Status (2025) | Key Findings |
---|---|---|---|---|
VLS-101 | ADC (MMAE) | Richter syndrome, NSCLC, TNBC | Phase I/II | 67% complete remission in RS-PDX models |
CS5001 | ADC (PNU) | Mantle cell lymphoma, Breast cancer | Phase I | Tumor regression in xenografts |
NVG-111 | BiTE | CLL, Solid tumors | Phase I | T-cell-mediated cytotoxicity |
Direct Targeting: Antibodies like UC-961 bind ROR1, inducing internalization and lysosomal degradation .
ADC Payload Delivery: Conjugated toxins (e.g., MMAE) disrupt microtubules, causing mitotic arrest .
Immune Activation: Bispecific antibodies (BiTEs) engage CD3⁺ T cells for tumor lysis .
Antigen Heterogeneity: Low ROR1 expression (<30% cells) in tumors like HCC limits ADC efficacy due to reduced bystander effects .
Toxicity Risks: Off-target binding to fetal tissues remains a concern despite ROR1’s absence in most adult tissues .
Resistance Mechanisms: Upregulation of MDR1 and BCL-2 in ROR1-knockdown cells necessitates combination therapies .
Next-Gen ADCs: Site-specific conjugation (e.g., CS5001’s PNU-159682) improves therapeutic index .
Combinatorial Regimens: Pairing with PD-1 inhibitors or BH3 mimetics to overcome resistance .
Diagnostic Expansion: Quantitative ELISA kits using antibody pairs (e.g., MP51139-1) for ROR1 detection in liquid biopsies .
The ROR1 recombinant monoclonal antibody is meticulously synthesized to ensure exceptional quality and specificity. The process begins with the isolation of B cells from an immunized animal, using the recombinant human ROR1 protein as the immunogen. Total RNA is extracted from these B cells and converted into cDNA through reverse transcription. The ROR1 antibody genes are then amplified using specific primers designed for the antibody constant regions and inserted into an expression vector. Following transfection, this vector is introduced into host cells, enabling the production of the ROR1 recombinant monoclonal antibody. After cell culture, the antibody is harvested from the supernatant and purified using affinity chromatography, resulting in a highly purified form suitable for various applications. Rigorous characterization assays, including ELISA and FC analysis, are performed to validate the antibody's specificity and functionality in detecting human ROR1 protein.
ROR1 exhibits very low kinase activity in vitro and is unlikely to function as a tyrosine kinase in vivo. It acts as a receptor for the ligand WNT5A, which activates downstream NFkB signaling pathways. This activation may lead to the inhibition of WNT3A-mediated signaling. In the inner ear, ROR1 plays a crucial role in the ability of spiral ganglion neurons to innervate auditory hair cells.
ROR1 is a transmembrane protein in the receptor tyrosine kinase family involved in intercellular signal communication and intracellular signal transduction. It regulates cell proliferation, differentiation, and metastasis and is considered a pseudokinase that promotes cancer cell survival. The significance of ROR1 as a therapeutic target stems from its differential expression pattern: it is predominantly expressed during embryogenesis in neural crest cells and shows minimal expression in normal adult tissues (limited to adipocytes and some B-cell precursors), while being highly expressed in various malignancies including chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL), lung adenocarcinoma, breast cancer, melanoma, renal cancer, gastric cancer, and colorectal cancer . This expression profile makes ROR1 an attractive target for antibody-based therapeutic strategies with potentially fewer off-target effects compared to targets that are widely expressed in normal tissues .
The production of ROR1 recombinant monoclonal antibodies involves a sophisticated multi-step process:
Immunization: Animals (typically mice or rabbits) are immunized with recombinant human ROR1 protein to generate an immune response .
B-cell isolation: B cells producing antibodies against ROR1 are isolated from the immunized animal .
Genetic engineering: Total RNA is extracted from these B cells and converted to cDNA through reverse transcription. The antibody genes are then amplified using specific primers designed for the antibody constant regions .
Vector construction: The amplified antibody genes are inserted into an expression vector .
Transfection and expression: The expression vector is introduced into host cells (commonly CHO or HEK293 cells) for antibody production .
Harvest and purification: The antibody is harvested from the cell culture supernatant and purified using affinity chromatography (typically Protein A) .
Characterization: The purified antibody undergoes rigorous characterization, including ELISA and flow cytometry analysis, to confirm its specificity and functionality .
For humanized antibodies, additional steps include CDR (complementarity-determining region) grafting, where the antigen-binding regions from the mouse or rabbit antibody are transferred to a human antibody framework to reduce immunogenicity in human patients .
ROR1 is expressed as a glycoprotein containing several distinct structural domains that can be targeted by monoclonal antibodies:
Extracellular region:
Immunoglobulin (Ig)-like domain
Frizzled domain
Kringle domain
Transmembrane region
Intracellular region:
Most therapeutic antibodies target the extracellular domains of ROR1, as these are accessible on the cell surface. Different antibodies may recognize epitopes in different domains, affecting their binding characteristics and functional properties. For instance, antibodies targeting the immunoglobulin-like domain might have different effects than those targeting the frizzled or kringle domains . The extracellular fragment (amino acids 30-406) is commonly used as an immunogen for antibody production . A diverse panel of antibodies targeting different epitopes across all three extracellular domains has been developed to explore various therapeutic approaches .
Researchers employ multiple complementary techniques to thoroughly characterize anti-ROR1 antibodies:
Flow Cytometry (FC):
Enzyme-Linked Immunosorbent Assay (ELISA):
Surface Plasmon Resonance (SPR) Spectroscopy:
Western Blotting:
Immunohistochemistry:
A comprehensive evaluation should include both cell-based and protein-based assays to ensure the antibody recognizes ROR1 in its native conformation as well as in denatured states, depending on the intended application .
Assessment of anti-ROR1 antibodies' functional effects requires a multi-faceted approach:
In vitro cellular assays:
Proliferation assays: MTT, XTT, or BrdU incorporation to measure growth inhibition
Apoptosis assays: Annexin V/PI staining, caspase activation, TUNEL assay
Migration and invasion assays: Wound healing, transwell, and Matrigel invasion assays
Signaling pathway analysis: Western blotting for downstream effectors (AKT, ERK) to determine if antibodies block ROR1-mediated signaling
Effector function evaluation:
ADCC (Antibody-Dependent Cellular Cytotoxicity): Co-culture of target cells with NK cells or PBMCs to assess antibody-mediated killing
CDC (Complement-Dependent Cytotoxicity): Incubation with complement to evaluate complement-mediated lysis
Internalization studies: Flow cytometry or confocal microscopy to assess antibody-induced receptor internalization
In vivo models:
RNAi comparison studies:
These assays should be performed with appropriate controls, including isotype-matched control antibodies and ROR1-negative cell lines, to confirm specificity of the observed effects .
For optimal results when using anti-ROR1 antibodies in flow cytometry, researchers should follow these methodological guidelines:
Sample preparation:
For cell lines: Harvest cells in exponential growth phase using enzyme-free dissociation buffer to preserve surface antigens
For primary samples: Isolate mononuclear cells using density gradient centrifugation
Wash cells in cold PBS containing 2% FBS or BSA (FACS buffer)
Antibody dilution:
Staining procedure:
Use 1-5 × 10^5 cells per sample
Perform Fc receptor blocking (especially for primary samples)
Incubate with primary antibody for 30-45 minutes at 4°C in the dark
Wash twice with FACS buffer
For unconjugated antibodies, add appropriate fluorochrome-labeled secondary antibody
Controls:
Analysis considerations:
Gate on viable cells using appropriate viability dye
Analyze using appropriate laser and filter settings for the fluorochrome
Quantify results as percent positive cells and mean/median fluorescence intensity
Troubleshooting:
If signal is weak, reduce washing steps or increase antibody concentration
High background may require additional blocking or reduced antibody concentration
For multi-color panels, perform compensation using single-stained controls
The dose or concentration should be optimized experimentally in dose-response or titration experiments for each specific application and cell type .
Different humanized anti-ROR1 antibodies exhibit distinct characteristics based on their epitope specificity:
The functional disparities between these antibodies highlight important considerations:
Epitope-function relationships: Antibodies targeting different domains of ROR1 can exhibit dramatically different functional properties, even with similar binding affinities.
Internalization dynamics: Antibodies with slower internalization rates may be more effective for ADCC but less optimal for antibody-drug conjugates that require internalization.
Effector functions: Most anti-ROR1 antibodies show limited natural cytotoxicity (ADCC/CDC), suggesting they may be better suited for armed approaches (ADCs, immunotoxins) rather than as naked antibodies .
Species cross-reactivity: Some antibodies bind to both human and mouse ROR1, allowing for more relevant preclinical studies, while others are human-specific .
These comparisons suggest that researchers should carefully select anti-ROR1 antibodies based on their intended application and mechanism of action rather than solely on binding affinity .
Developing effective ROR1-targeted therapeutic modalities requires careful consideration of several critical factors:
Target density and accessibility:
Internalization kinetics:
Antibody engineering considerations:
Payload selection for ADCs:
Match payload potency to target density
Consider bystander effect potential based on tumor architecture
Evaluate payload stability in circulation versus release kinetics
For CAR-T approaches:
Optimal epitope selection to avoid steric hindrance
Appropriate CAR design (costimulatory domains, hinge length)
Strategies to mitigate on-target/off-tumor toxicity in adipose tissue
Combination strategies:
Biomarker development:
Methods for patient selection based on ROR1 expression levels
Pharmacodynamic markers to assess target engagement
Resistance monitoring strategies
The relatively modest cytotoxicity observed with naked anti-ROR1 antibodies suggests that most clinical development should focus on armed antibodies or cell-based approaches like CAR-T cells to maximize therapeutic potential .
Addressing safety concerns for anti-ROR1 therapeutics requires comprehensive preclinical assessment and strategic approaches:
Thorough tissue cross-reactivity studies:
Immunohistochemistry screening across a panel of normal human tissues
Special attention to tissues with reported low-level ROR1 expression:
Both frozen and fixed tissues should be evaluated to account for potential epitope masking
Dosing strategy optimization:
Careful dose escalation to identify therapeutic window
Alternative dosing schedules to mitigate toxicity
Exploration of fractionated dosing approaches
Sophisticated preclinical models:
Humanized mouse models expressing human ROR1 in normal tissues
Non-human primate studies for antibodies with cross-reactivity
Patient-derived xenografts to better predict efficacy/toxicity balance
Engineering approaches to enhance safety:
Bispecific antibodies requiring dual antigen recognition
Masked antibodies that activate only in the tumor microenvironment
Switchable CAR-T systems with titratable activity
Clinical trial design considerations:
Inclusion of robust safety monitoring:
Adipose tissue function assessments
B-cell development markers
Lung function tests
Implementation of risk mitigation strategies:
Prompt intervention protocols
Predetermined dose modification guidelines
Biomarker-guided patient selection
Comparative safety assessment:
The selective expression pattern of ROR1 provides a favorable therapeutic window, but meticulous safety evaluation remains essential, particularly for highly potent modalities like ADCs and CAR-T cells where even low-level expression in normal tissues could lead to toxicity .
Accurate detection and quantification of ROR1 present several technical challenges that can be addressed through methodological refinements:
Optimizing antibody selection:
Enhancing sensitivity for low-expressing samples:
Standardization and quantification approaches:
Use calibrated flow cytometry with antibody-binding capacity (ABC) beads
Implement quantitative PCR with validated primers and probes
Develop standard curves with recombinant ROR1 protein
Include quantitative controls (cell lines with known ROR1 expression levels)
Multi-parametric analysis:
Combine ROR1 detection with lineage markers
Correlate protein expression with mRNA levels
Consider single-cell approaches for heterogeneous samples
Sample preparation considerations:
For tissue samples:
Optimize fixation time to preserve epitopes
Consider antigen retrieval methods
For flow cytometry:
Use enzyme-free dissociation methods
Process samples rapidly to prevent receptor internalization
Negative and positive controls:
Include ROR1 knockdown/knockout cells as negative controls
Use engineered ROR1-overexpressing cells as positive controls
Compare results with established ROR1-positive cancer cell lines
These methodological refinements can significantly improve the reliability of ROR1 detection and quantification, particularly in samples with heterogeneous or low-level expression .
When faced with discrepancies between results obtained using different anti-ROR1 antibody clones, researchers should implement a systematic troubleshooting and reconciliation approach:
Epitope mapping analysis:
Technical validation comparison:
Review the validation parameters for each antibody:
Specificity testing methodology
Positive and negative controls used
Applications for which each clone is validated (FC, IHC, WB)
Compare detection methods and sensitivity limits
Orthogonal confirmation approaches:
Validate findings using non-antibody-based methods:
mRNA expression (RT-PCR, RNA-seq)
CRISPR knockout controls
Recombinant expression systems
Use multiple antibodies targeting different epitopes in parallel
Biological context interpretation:
Consider protein isoforms or splice variants
Evaluate potential proteolytic processing of ROR1
Assess glycosylation status, which may affect epitope recognition
Examine microenvironmental factors that could influence expression or accessibility
Reconciliation framework:
Create a decision matrix weighing evidence from multiple assays
Prioritize results from the most rigorously validated antibodies
Consider the biological relevance of each detection method
Design follow-up experiments to specifically address discrepancies
Reporting recommendations:
Transparently document all antibodies used (clone, vendor, catalog number)
Specify exact experimental conditions for each antibody
Acknowledge limitations and discrepancies in publications
Provide raw data when possible to allow independent interpretation
Rigorous quality control is essential when selecting anti-ROR1 antibodies for research applications:
Manufacturing quality metrics:
Specificity validation:
Positive controls: ROR1-overexpressing cell lines
Negative controls:
ROR1 knockout/knockdown cells
Non-expressing cell lines
Cross-reactivity assessment with related proteins (especially ROR2)
Absorption/competition studies with recombinant ROR1
Application-specific performance parameters:
For Flow Cytometry:
Signal-to-noise ratio on positive vs. negative cells
Staining index calculation
Optimal working concentration determination (titration series)
Performance with fixed vs. live cells
For Western Blotting:
Minimal non-specific bands
Performance under reducing vs. non-reducing conditions
Sensitivity (minimum detectable concentration)
For Immunohistochemistry:
Staining pattern consistency with known biology
Background levels in negative tissues
Performance across different fixation methods
Antigen retrieval requirements
Functional characterization:
Ability to detect native vs. denatured protein
Epitope accessibility in different contexts
Internalization properties if relevant to application
Functional effects (neutralizing vs. non-neutralizing)
Stability parameters:
A standardized validation protocol should be established for each application, incorporating positive and negative controls and reference standards to ensure reliable, reproducible results across experiments .
Advanced engineering approaches are expanding the therapeutic potential of anti-ROR1 antibodies:
Antibody-Drug Conjugates (ADCs):
Conjugation of cytotoxic payloads to anti-ROR1 antibodies
Strategic selection of linkers based on internalization kinetics
Payload selection matched to ROR1 expression level
Site-specific conjugation for improved homogeneity and stability
Bispecific antibody formats:
ROR1 × CD3 bispecifics to redirect T cells to ROR1+ tumors
ROR1 × NK cell receptor bispecifics for enhanced ADCC
Dual targeting of ROR1 with complementary tumor antigens to improve specificity
Conditional activation designs requiring dual antigen recognition
Fc engineering strategies:
CAR-T cell and cellular therapy applications:
Optimization of anti-ROR1 scFv fragments for CAR construction
Novel CAR designs with customized costimulatory domains
Logic-gated CAR systems requiring multiple antigen recognition
Integration with gene editing to enhance persistence and efficacy
Novel antibody formats:
Single-domain antibodies for improved tissue penetration
Tribodies and other multivalent formats for avidity enhancement
Intrabodies targeting intracellular domains of ROR1
Masked antibodies activated by tumor-specific proteases
Combination therapy designs:
Synergistic targeting of ROR1 signaling partners
Incorporation into bi- or tri-specific immune checkpoint inhibitors
Rational combinations with standard chemotherapy regimens
These engineering approaches aim to overcome the limitations observed with naked anti-ROR1 antibodies, particularly addressing the relatively low natural cytotoxicity and moderate expression levels of ROR1 in tumor cells .
Antibody-based research has uncovered several important aspects of ROR1 biology with therapeutic implications:
Signaling pathway discoveries:
Expression pattern insights:
Dramatic overexpression in multiple cancer types including:
Refined understanding of normal tissue expression:
Functional roles in cancer biology:
Therapeutic vulnerability insights:
Antibodies blocking different epitopes revealed domain-specific functions
Differential responses across cancer types suggest context-dependent roles
Varying internalization rates of antibody-ROR1 complexes revealed dynamic receptor trafficking
Biomarker potential:
Correlation between ROR1 expression and clinical outcomes
Association with specific cancer subtypes
Potential for patient stratification in clinical trials
Developmental biology connections:
These insights provide important context for therapeutic development and suggest potential mechanisms of action and resistance that should be considered when designing anti-ROR1 targeted therapies .
Emerging evidence suggests several promising combination approaches with anti-ROR1 antibodies:
Combinations with pathway inhibitors:
Wnt pathway inhibitors: Since Wnt-5a is a candidate ligand for ROR1, combining anti-ROR1 antibodies with Wnt pathway inhibitors may provide synergistic effects by blocking both the receptor and its signaling pathway
PI3K/AKT inhibitors: Given ROR1's role in AKT activation through TCL1 co-activation, this combination could enhance apoptotic responses
BTK inhibitors: For hematological malignancies, particularly CLL and MCL, this combination has shown promise in preclinical models
Immunotherapy combinations:
Immune checkpoint inhibitors: Combining anti-ROR1 antibodies with anti-PD-1/PD-L1 or anti-CTLA-4 antibodies to enhance immune recognition and elimination of tumor cells
Bispecific approaches: Dual-targeting of ROR1 and immune activators (CD3, 4-1BB) in a single molecule
Cellular therapy enhancements: Anti-ROR1 antibodies alongside CAR-T cell approaches targeting different antigens
Combinations with conventional therapies:
Chemotherapy: Humanized anti-ROR1 antibodies (h1B8 and h6D4) could sensitize tumor cells to standard chemotherapeutic agents
Radiation therapy: Potential radiosensitizing effects through disruption of survival pathways
Targeted therapies: Combinations with EGFR, HER2, or other targeted agents based on tumor type
Multi-antibody approaches:
Cocktails of anti-ROR1 antibodies: Targeting different epitopes simultaneously may enhance efficacy
Sequential antibody therapy: Adapting treatment based on evolving resistance mechanisms
Complementary target pairs: Combining ROR1 targeting with antibodies against synergistic targets like CD20 for B-cell malignancies
Enhanced antibody formats:
ADC combinations: Lower doses of multiple ADCs targeting different antigens to reduce toxicity while maintaining efficacy
Tribody approaches: Creating molecules that simultaneously target ROR1, a tumor antigen, and an immune cell receptor
Initial evidence from humanized anti-ROR1 antibodies (h1B8 and h6D4) demonstrates substantial anti-tumor activity in multiple cancer models, including lung cancer xenografts, c-Myc/Alb-cre liver cancer transgenic mice, and MMTV-PyMT breast cancer mice . These findings provide a strong rationale for further exploration of combination strategies to enhance therapeutic outcomes across various cancer types.
Researchers working with ROR1 recombinant monoclonal antibodies should consider several critical factors to ensure robust and meaningful results:
Antibody selection and validation:
Experimental design considerations:
Account for ROR1's relatively low expression level compared to other targets
Optimize antibody concentrations through careful titration (1:50-1:200 range for flow cytometry)
Include parallel experiments with ROR1 knockdown/knockout to confirm specificity
Design time-course studies to capture dynamic responses
Technical optimization:
Interpretative frameworks:
Consider ROR1's biological context in your experimental system
Account for potential off-target effects
Interpret results in light of ROR1's known signaling pathways
Recognize limitations of in vitro systems versus in vivo complexity
Translational relevance:
Design experiments that address clinically relevant questions
Consider pharmacokinetic and pharmacodynamic parameters when applicable
Include appropriate model systems that recapitulate human disease features
Develop biomarkers that could translate to clinical applications
By carefully addressing these considerations, researchers can maximize the utility of ROR1 recombinant monoclonal antibodies in advancing our understanding of ROR1 biology and developing novel targeted therapies for ROR1-expressing malignancies .
The field of ROR1-targeted antibody research is poised for significant advances in several key areas:
Next-generation antibody technologies:
Development of proteolysis-targeting chimeras (PROTACs) incorporating anti-ROR1 antibodies
Application of switchable CAR systems using anti-ROR1 antibody fragments
Integration with nanobody platforms for enhanced tissue penetration
Novel scaffold approaches beyond traditional antibody formats
Advanced therapeutic modalities:
Optimized antibody-drug conjugates with improved therapeutic index
Tri-specific antibodies targeting ROR1 alongside complementary targets
Cell-penetrating antibodies addressing intracellular ROR1 functions
Radioimmunotherapy approaches using anti-ROR1 antibodies
Precision medicine applications:
Development of companion diagnostics using anti-ROR1 antibodies
Biomarker stratification approaches for patient selection
Real-time monitoring of treatment response and resistance
Liquid biopsy applications for circulating tumor cells
Manufacturing and development innovations:
Continued improvements in humanization and deimmunization strategies
Enhanced production platforms for more consistent recombinant antibodies
Cost-effective manufacturing approaches to improve accessibility
Streamlined regulatory pathways for antibody-based therapeutics
Expanded disease applications:
Beyond the current focus on CLL, MCL, and solid tumors
Exploration of ROR1's role in cancer stem cells and minimal residual disease
Investigation of non-oncology applications based on developmental biology insights
Potential applications in fibrosis or other pathologies with aberrant Wnt signaling
The continued development of humanized anti-ROR1 antibodies with enhanced specificity, affinity, and functional properties will drive clinical translation, with several candidates already showing promising preclinical efficacy . The clinical progress of Zilovertamab (UC-961) provides important validation for the ROR1-targeting approach and will inform future antibody development efforts .
Emerging technologies in antibody engineering and production are poised to transform anti-ROR1 therapeutic development:
AI-driven antibody design:
Computational prediction of optimal complementarity-determining regions (CDRs)
Structure-based epitope mapping to target functionally critical domains
In silico affinity maturation to enhance binding properties
Prediction of developability characteristics (stability, manufacturability)
Site-specific conjugation technologies:
Engineered cysteine residues for controlled conjugation sites
Incorporation of non-natural amino acids for click chemistry
Enzymatic approaches for site-specific modification
These advances will create more homogeneous ADCs with improved therapeutic index
Advanced expression systems:
Glycoengineered host cells for controlled glycosylation profiles
Continuous manufacturing platforms for more consistent product quality
Cell-free expression systems for rapid prototyping
Improved purification technologies for higher yields and purity
Novel antibody formats:
Smaller antibody fragments with enhanced tissue penetration
Multi-specific formats targeting ROR1 alongside complementary pathways
pH-sensitive binding domains for conditional activation
Intracellular antibody fragments delivered via novel technologies
Manufacturing innovations:
Single-use bioreactor systems for flexible production
Process analytical technologies for real-time quality monitoring
Automated purification strategies to reduce variability
Streamlined development pathways from discovery to GMP production
These technological advances will address current limitations of anti-ROR1 antibodies:
Improving binding characteristics to overcome the relatively low ROR1 expression levels
Enhancing functional properties through optimized Fc engineering
Creating more consistent batches with reduced immunogenicity risk
Enabling more sophisticated targeting strategies with multi-functional antibodies