ROR1 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Significance

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 .

Development and Production

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 .

Preclinical Findings

  • 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% .

Clinical Applications and Trials

ROR1-targeted antibody-drug conjugates (ADCs) and immunotherapies are under evaluation:

Table 2: Clinical-Stage ROR1-Targeted Therapies

TherapyTypeTarget IndicationsStatus (2025)Key Findings
VLS-101ADC (MMAE)Richter syndrome, NSCLC, TNBCPhase I/II67% complete remission in RS-PDX models
CS5001ADC (PNU)Mantle cell lymphoma, Breast cancerPhase ITumor regression in xenografts
NVG-111BiTECLL, Solid tumorsPhase IT-cell-mediated cytotoxicity

Mechanism of Action

  • 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 .

Challenges and Limitations

  • 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 .

Future Directions

  • 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 .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

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.

Form
Liquid
Lead Time
Typically, we can ship products within 1-3 working days after receiving your orders. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timelines.
Synonyms
dJ537F10.1; Inactive tyrosine protein kinase transmembrane receptor ROR1; MGC99659; Neurotrophic tyrosine kinase; Neurotrophic tyrosine kinase receptor related 1; Neurotrophic tyrosine kinase; receptor related 1; NTRKR1; OTTHUMP00000010573; OTTHUMP00000010574; OTTMUSP00000008344; Receptor tyrosine kinase like orphan receptor 1; receptor-related 1; RGD1559469; ROR 1; ROR1; ROR1_HUMAN; RP11 24J23.1; Tyrosine kinase like orphan receptor 1; Tyrosine protein kinase transmembrane receptor ROR1; Tyrosine-protein kinase transmembrane receptor ROR1
Target Names
Uniprot No.

Target Background

Function

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.

Gene References Into Functions
  1. This study found that in scid hu mice, ROR1 was highly expressed in a proportion of bone marrow, spleen, and blood B cells, primarily immature B cells. PMID: 29850623
  2. Results indicate that ROR1 is the direct target of miR30a and demonstrate that ROR1 contributes to mir30a-mediated suppression of TNBC (triple negative breast cancer) cell invasion and migration. PMID: 29693179
  3. ROR1 and ROR2 play distinct roles in endometrial cancer. ROR1 may promote tumor progression, while ROR2 may act as a tumor suppressor in endometrioid endometrial cancer. PMID: 29395309
  4. ROR1 is a promising immunotherapeutic target in many epithelial tumors; however, high cell surface ROR1 expression in multiple normal tissues raises concerns about on-target off-tumor toxicities. The clinical translation of ROR1-targeted therapies necessitates careful monitoring of toxicities to normal organs and may require strategies to ensure patient safety. PMID: 27852699
  5. This report shows that ROR1 is highly expressed in colorectal cancer (CRC) tissues compared to their adjacent normal tissues. The Kaplan-Meier curve indicated that CRC patients with higher ROR1 expression had significantly shorter overall survival (OS), while those with lower ROR1 expression had longer OS. PMID: 28427197
  6. This study reveals that 14-3-3zeta plays a critical role in Wnt5a/ROR1 signaling, leading to enhanced CLL migration and proliferation. PMID: 28465528
  7. These studies indicate that HS1 plays an important role in ROR1-dependent Wnt5a-enhanced chemokine-directed leukemia-cell migration. PMID: 28465529
  8. This study demonstrates the expression of ROR1 and its putative ligand Wnt5a in Ewing sarcomas, and of an active ROR1 protein variant in cell lines. ROR1 silencing impaired cell migration in vitro. PMID: 26739507
  9. The expression of ROR1 may promote leukemia-cell activation and survival, and enhance disease progression in patients with chronic lymphocytic leukemia. PMID: 27815263
  10. This study focuses on the mechanistic regulation and linkage of the ROR1-HER3 and Hippo-YAP pathway in a cancer-specific context. PMID: 28114269
  11. Data show that ROR1 contributes to melanoma progression by promoting cell growth and migration. PMID: 26509654
  12. High ROR1-DNAJC6 expression is associated with neoplasms. PMID: 27153396
  13. This review presents an overview of the efforts and scientific achievements in targeting the ROR family, particularly ROR-1, for the diagnosis and treatment of chronic lymphocytic leukemia. PMID: 28160756
  14. This study suggests that strong ROR1 expression might be an independent adverse prognostic factor in triple negative breast cancer. PMID: 26874851
  15. Ror1 is crucial for spiral ganglion neurons to innervate auditory hair cells. Impairment of ROR1 function significantly affects the development of the inner ear and hearing in humans and mice. PMID: 27162350
  16. Targeting ROR1 can induce differentiation of cancer stem cells and inhibit metastasis in glioblastoma. Additionally, ROR1 may serve as a potential marker for glioblastoma stem cells and a potential target for glioblastoma stem cell therapy. PMID: 26923195
  17. The expression of ROR1 was significantly higher in colorectal carcinoma tissues than in tumor-adjacent tissues. PMID: 27126945
  18. These findings revealed that miR382 inhibits migration and invasion by targeting ROR1 through regulating EMT in ovarian cancer, and might serve as a tumor suppressor in ovarian cancer. PMID: 26575700
  19. Data show that silencing receptor tyrosine kinases (RTKs) ROR2 and ROR1 has a strong inhibitory effect on the ability of ovarian cancer cells to proliferate, migrate and invade. PMID: 26515598
  20. The b-catenin-independent WNT score correlated with reduced overall survival only in the metastasized situation. This aligns with in vitro results regarding the alternative WNT receptors ROR1 and ROR2, which promote invasion. PMID: 26862065
  21. The present findings support the notion that ROR1 sustains lung adenocarcinoma survival, at least in part, through direct physical interaction with ASK1. PMID: 26661061
  22. High expression of ROR1 (63%), pAkt (36%), and pCREB (20%) was observed in gastric adenocarcinomas, and expression of these proteins was well intercorrelated. PMID: 26245996
  23. This study identifies an interaction between ROR1 and ROR2 that is required for Wnt5a signaling, which promotes leukemia chemotaxis and proliferation. PMID: 26690702
  24. miR-27b-3p suppresses cell proliferation through targeting receptor tyrosine kinase like orphan receptor 1 in gastric cancer. PMID: 26576539
  25. Data show that the majority of chronic lymphocytic leukemia (CLL) patients had antibodies against receptor tyrosine kinase ROR1. PMID: 26562161
  26. This study reports an unexpected function of ROR1 as a scaffold of cavin-1 and caveolin-1, two essential structural components of caveolae. PMID: 26725982
  27. Both IGF1R and ROR1 can be effectively targeted by SB modified CAR T cells. PMID: 26173023
  28. Significantly down-regulates the activity of the PI3K/AKT/mTOR signaling pathway. PMID: 25978653
  29. Human ROR1 and ROR2 are receptor tyrosine kinase-like pseudokinases. PMID: 25029443
  30. The newly developed OSU-2S delivery using ROR1-directed immunonanoparticles provide selective targeting of OSU-2S to MCL and other ROR1(+) malignancies, sparing normal B cells. PMID: 25937048
  31. ROR1 expression is correlated with malignant attributes of ovarian cancer and it may serve as a novel prognostic marker in ovarian cancer. PMID: 25056203
  32. Data indicate that shRNA silencing of type I receptor tyrosine kinase-like orphan receptor (ROR1) cells, or treatment with anti-ROR1 mAb UC-961 impaired the capacity of ovarian cancer cells to form spheroids or tumor xenografts. PMID: 25411317
  33. ROR1 is only detectable in embryonic tissue and generally absent in adult tissue, making the protein an ideal drug target for cancer therapy. [Review] PMID: 24752542
  34. CLL cells expressed different isoforms of ROR1. PMID: 24205204
  35. Data shows that ROR1 and ROR2 are inversely expressed in melanomas and negatively regulate each other. Also, hypoxia initiates a shift of ROR1-positive melanomas to a more invasive, ROR2-positive phenotype. PMID: 24104062
  36. ROR1 can interact with TCL1 and enhance leukemogenesis in Emu-TCL1 transgenic mice. PMID: 24379361
  37. Our results show that customizing spacer design and increasing affinity of ROR1-CARs enhances T-cell effector function and recognition of ROR1(+) tumors. PMID: 23620405
  38. ROR1 may play a role in the survival of melanoma cells. PMID: 23593420
  39. Data indicate that type I receptor tyrosine kinase-like orphan receptor ROR1 may regulate EMT and metastasis, and that antibodies targeting ROR1 can inhibit cancer progression and metastasis. PMID: 23771907
  40. The receptor tyrosine kinase ROR1 was overexpressed in most patients with various hematological malignancies of both lymphoid and myeloid origins. PMID: 22988987
  41. Cell surface expression in pediatric B-ALL along with its virtual absence from normal tissues and circulating cells makes ROR1 a promising target for mAb-based therapies. PMID: 23285131
  42. Many different human cancers express ROR1, and ROR1 may play a functional role in promoting tumor cell growth. PMID: 23041612
  43. t(1;19) Acute Lymphoblastic Leukemia cells universally exhibit expression of and dependence on the cell surface receptor ROR1. PMID: 23153538
  44. ROR1 was overexpressed in acute lymphoblastic leukemia. PMID: 22369092
  45. ROR1 is expressed in human breast cancers and has biological and clinical significance. PMID: 22403610
  46. Nuclear-localized ROR1 may play an important role in cell migration and cytoskeleton remodeling. PMID: 22199287
  47. Ror1 undergoes complex post-translational modifications by glycosylation and mono-ubiquitination. These modifications regulate Ror1 localization and signaling, and are highly variable among individual chronic lymphocytic leukemia patients. PMID: 21481194
  48. ROR1 is uniformly and highly expressed in all chronic lymphocytic leukemia (CLL) cases at initial diagnosis and can serve as a diagnostic tool. PMID: 21531460
  49. ROR1 is expressed on hematogones (non-neoplastic human B-lymphocyte precursors) and a minority of precursor-B acute lymphoblastic leukemia. PMID: 21813176
  50. A panel of mAbs demonstrated high affinity and specificity for a diverse set of epitopes that involve all three extracellular domains of ROR1, are accessible on the cell surface, and mediate internalization. PMID: 21698301

Show More

Hide All

Database Links

HGNC: 10256

OMIM: 602336

KEGG: hsa:4919

STRING: 9606.ENSP00000360120

UniGene: Hs.128753

Involvement In Disease
Deafness, autosomal recessive, 108 (DFNB108)
Protein Families
Protein kinase superfamily, Tyr protein kinase family, ROR subfamily
Subcellular Location
Membrane; Single-pass type I membrane protein. Cell projection, axon.
Tissue Specificity
Expressed strongly in human heart, lung and kidney, but weakly in the CNS. Isoform Short is strongly expressed in fetal and adult CNS and in a variety of human cancers, including those originating from CNS or PNS neuroectoderm.

Q&A

What is ROR1 and why is it a significant target for monoclonal antibodies in cancer research?

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 .

How are ROR1 recombinant monoclonal antibodies typically produced?

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 .

What are the main structural domains of ROR1 that can be targeted by monoclonal antibodies?

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:

    • Tyrosine kinase domain

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 .

What experimental methods are used to evaluate the specificity and affinity of anti-ROR1 antibodies?

Researchers employ multiple complementary techniques to thoroughly characterize anti-ROR1 antibodies:

  • Flow Cytometry (FC):

    • Evaluates binding to ROR1-expressing cells versus control cells

    • Typically used at dilutions of 1:50-1:200

    • Quantifies cell surface expression levels and antibody binding

  • Enzyme-Linked Immunosorbent Assay (ELISA):

    • Measures direct binding to recombinant ROR1 protein

    • Determines specificity by testing cross-reactivity with related proteins

    • Enables affinity determination through titration experiments

  • Surface Plasmon Resonance (SPR) Spectroscopy:

    • Provides precise measurements of binding kinetics (ka and kd rates)

    • Calculates equilibrium dissociation constant (KD) to quantify affinity

    • Allows real-time observation of antibody-antigen interactions

  • Western Blotting:

    • Confirms specificity by detecting ROR1 protein at the expected molecular weight (~101 kDa)

    • Assesses potential cross-reactivity with other proteins

  • Immunohistochemistry:

    • Evaluates binding to ROR1 in tissue samples from various tumors and normal tissues

    • Confirms specificity in the context of complex tissue environments

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 .

How can researchers assess the functional effects of anti-ROR1 antibodies in cancer models?

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:

    • Xenograft models: Human tumor cells implanted in immunodeficient mice

    • Transgenic models: Including:

      • c-Myc/Alb-cre liver cancer transgenic mice

      • MMTV-PyMT breast cancer mice

    • Parameters measured: Tumor growth, survival, metastasis, pharmacokinetics, toxicity

  • RNAi comparison studies:

    • Using ROR1 knockdown by shRNA as a positive control to compare antibody efficacy

    • Helps validate that observed effects are specifically due to ROR1 targeting

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 .

What are the recommended protocols for using anti-ROR1 antibodies in flow cytometry?

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:

    • Start with the recommended dilution range of 1:50-1:200

    • Perform titration experiments to determine optimal concentration

    • Prepare antibody in appropriate buffer (PBS, pH 7.4, without preservatives recommended)

  • 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:

    • Include unstained cells for autofluorescence

    • Use isotype control (e.g., Anti-HEL Human IgG1(E356D/M358L)-Kappa for Zilovertamab)

    • Include both ROR1-positive and ROR1-negative cell lines as biological 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 .

How do different humanized anti-ROR1 antibodies compare in terms of epitope specificity and functional properties?

Different humanized anti-ROR1 antibodies exhibit distinct characteristics based on their epitope specificity:

AntibodyEpitope LocationAffinity (KD)Internalization RateADCC ActivityCDC ActivityIn vivo EfficacyReference
h1B8Not specifiedHigh affinityNot specifiedDemonstratedNot specifiedEffective in lung, liver, and breast cancer models
h6D4Not specifiedHigh affinityNot specifiedDemonstratedNot specifiedEffective in lung, liver, and breast cancer models
Zilovertamab (UC-961)Not fully disclosedNot specifiedLong plasma half-lifeBlocks ROR1 signalingNot specifiedInhibits engraftment of leukemia cells
Panel from Yang et al.Diverse epitopes across all 3 extracellular domainsHighest affinity mAb had slowest internalizationSlow to moderateWeak, only in highest affinity mAbNone observedNot specified

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 .

What are the key considerations when developing ROR1-targeted antibody-drug conjugates or immunotherapeutic approaches?

Developing effective ROR1-targeted therapeutic modalities requires careful consideration of several critical factors:

  • Target density and accessibility:

    • ROR1 has relatively low cell surface density compared to other cancer targets

    • This makes it potentially more suitable for armed antibody approaches (ADCs, immunotoxins) rather than naked antibodies

    • Quantitative assessment of ROR1 molecules per cell is crucial for predicting efficacy

  • Internalization kinetics:

    • For ADCs and immunotoxins, efficient internalization is essential

    • Different anti-ROR1 antibodies show varying rates of internalization

    • Selection of antibodies with appropriate internalization properties for the specific payload is critical

  • Antibody engineering considerations:

    • Format selection (IgG, Fab, scFv, bispecific)

    • Fc engineering to enhance or eliminate effector functions based on mechanism

    • Humanization strategy to minimize immunogenicity while preserving affinity

    • Linker chemistry and conjugation sites for ADCs

  • 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:

    • Potential synergy with Wnt pathway inhibitors

    • Combinations with immune checkpoint inhibitors

    • Sequential therapy approaches

  • 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 .

How can researchers address potential off-target effects and toxicity concerns with anti-ROR1 therapeutics?

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:

      • Adipocytes

      • Pancreatic tissue

      • B-cell precursors (hematogones)

      • Lung tissue

    • 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:

    • Benchmarking against Zilovertamab's clinical safety profile, which has shown "no discernable dose-limiting toxicity" in phase 1 studies

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 .

What strategies can overcome limitations in detecting and quantifying ROR1 expression in research samples?

Accurate detection and quantification of ROR1 present several technical challenges that can be addressed through methodological refinements:

  • Optimizing antibody selection:

    • Use multiple antibodies targeting different epitopes to confirm results

    • Select antibodies validated for specific applications (FC, IHC, WB)

    • Consider clone-specific performance characteristics:

      • Clone 2H6 works well for ELISA and Western blot

      • Certain clones may perform better for flow cytometry

  • Enhancing sensitivity for low-expressing samples:

    • Implement signal amplification techniques:

      • Tyramide signal amplification for IHC

      • Polymeric detection systems

    • Reduce background through optimized blocking:

      • Use species-specific sera

      • Include FcR blocking reagents for flow cytometry

    • Increase antibody concentration within validated ranges (1:50-1:200)

  • 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 .

How should researchers interpret conflicting results when using different anti-ROR1 antibody clones?

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:

    • Different antibodies target distinct epitopes on ROR1

    • Map the binding domains of each antibody (Ig-like, Frizzled, or Kringle domains)

    • Epitope accessibility may vary depending on:

      • Protein conformation

      • Post-translational modifications

      • Protein-protein interactions

  • 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

What quality control parameters should be evaluated when selecting and validating anti-ROR1 antibodies for specific research applications?

Rigorous quality control is essential when selecting anti-ROR1 antibodies for research applications:

  • Manufacturing quality metrics:

    • Purity level (>95% by SDS-PAGE is standard)

    • Endotoxin content (<1EU/mg, determined by LAL gel clotting assay)

    • Aggregation profile (by size-exclusion chromatography)

    • Batch-to-batch consistency documentation

  • 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:

    • Detection of expected molecular weight band (~101kDa)

    • 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:

    • Shelf-life data under recommended storage conditions

    • Freeze-thaw stability

    • Performance after reconstitution (2 weeks at 2-8°C is typical)

    • Temperature sensitivity during experimental procedures

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 .

How are ROR1 antibodies being engineered for enhanced therapeutic efficacy?

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:

    • Enhanced ADCC through afucosylation or amino acid substitutions

    • Extended half-life variants (E356D/M358L mutations)

    • Reduced immunogenicity through deimmunization

    • pH-dependent binding for improved recycling

  • 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 .

What novel insights about ROR1 biology have been revealed through antibody-based research approaches?

Antibody-based research has uncovered several important aspects of ROR1 biology with therapeutic implications:

  • Signaling pathway discoveries:

    • ROR1 functions as a pseudokinase that promotes cancer cell survival

    • Interaction with Wnt-5a induces signaling cascades

    • TCL1 co-activation with ROR1 enhances AKT signaling

    • ROR1 regulates cell proliferation, differentiation, and metastasis through complex signaling networks

  • Expression pattern insights:

    • Dramatic overexpression in multiple cancer types including:

      • Lung adenocarcinoma

      • Breast cancer

      • Melanoma

      • Renal cancer

      • Gastric cancer

      • Colorectal cancer

      • CLL and MCL (hematological malignancies)

    • Refined understanding of normal tissue expression:

      • Neural crest cells during embryogenesis

      • Low expression in adipocytes

      • Limited expression in pancreatic tissue

      • B-cell precursors (hematogones)

  • Functional roles in cancer biology:

    • RNAi studies paired with antibody approaches revealed ROR1's role in:

      • Cancer cell proliferation

      • Migration and invasion

      • Resistance to apoptosis

    • Targeting ROR1 with shRNAs significantly inhibits proliferation and migration of cancer cells

  • 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:

    • Role in organ morphogenesis during embryonic development

    • Function in nervous system development

    • Involvement in neural progenitor cell maintenance and survival

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 .

What are the most promising combination strategies involving anti-ROR1 antibodies for cancer treatment?

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.

What are the key considerations for researchers designing experiments with ROR1 recombinant monoclonal antibodies?

Researchers working with ROR1 recombinant monoclonal antibodies should consider several critical factors to ensure robust and meaningful results:

  • Antibody selection and validation:

    • Choose antibodies with validated specificity for the intended application

    • Consider epitope location and its relevance to research questions

    • Verify performance in your specific experimental system

    • Include appropriate positive and negative controls

  • 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:

    • Follow recommended storage conditions (2-8°C for short-term, -80°C for long-term)

    • Minimize freeze-thaw cycles to maintain antibody integrity

    • Use recommended buffers (PBS, pH 7.4, without preservatives)

    • Optimize protocols for specific cell lines or tissue types

  • 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 .

What future developments can we anticipate in the field of ROR1-targeted antibody research?

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 .

How might advances in antibody engineering and production technologies impact the development of next-generation anti-ROR1 therapeutics?

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

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.