CLDN18 monoclonal antibodies are humanized or chimeric IgG1 antibodies designed to bind CLDN18.2, a transmembrane protein critical for maintaining epithelial barrier integrity. CLDN18.2 is selectively expressed in gastric (58%), gastroesophageal junction (60%), and pancreatic (20%) adenocarcinomas, with minimal expression in healthy tissues, creating a therapeutic window .
Key characteristics of CLDN18.2:
Structural accessibility: Extracellular loops enable antibody binding .
Stability: Retained expression during metastasis, supporting systemic treatment .
Functional duality: May exhibit tumor-promoting or suppressive roles depending on context .
CLDN18 monoclonal antibodies drive antitumor activity through multiple mechanisms:
Antibody-dependent cellular cytotoxicity (ADCC): Recruits natural killer (NK) cells and macrophages to lysе CLDN18.2+ cells .
Complement-dependent cytotoxicity (CDC): Activates the complement cascade, inducing direct cell lysis .
Disruption of CLDN18.2-mediated signaling pathways inhibits proliferation and induces apoptosis in preclinical models .
Example: Givastomig (CLDN18.2×4-1BB bispecific antibody) localizes 4-1BB activation to the tumor microenvironment (TME), enhancing T-cell activity without systemic toxicity .
Key findings from in vitro and in vivo studies:
Zolbetuximab: Demonstrated 37% objective response rate (ORR) in phase II trials for CLDN18.2+ gastric cancer .
AB011: Phase I trial (NCT04400383) reported 66.7% disease control rate in pancreatic cancer .
SPOTLIGHT/GLOW trials: Zolbetuximab + chemotherapy improved progression-free survival (PFS) in metastatic gastric cancer .
| Therapy Type | Mechanism | Advantages | Limitations |
|---|---|---|---|
| Monoclonal Antibody | ADCC/CDC, signal inhibition | Broad applicability | Limited penetration in dense TME |
| CAR-T | Direct T-cell activation | Durable responses | Risk of cytokine release syndrome |
| Antibody-Drug Conjugate | Targeted payload delivery | High potency | Off-target toxicity concerns |
Numerous studies have highlighted the significant role of CLDN18 in various biological processes:
CLDN18.2 (Claudin-18.2) is a specific isoform of the Claudin-18 protein, which belongs to the claudin family of tight junction tetraspan cell surface proteins. It plays a critical role in maintaining cell-to-cell adhesion and regulating transport between adjacent cells in epithelial tissues . What makes CLDN18.2 particularly attractive as a therapeutic target is its highly restricted expression pattern in normal tissues coupled with significant overexpression in multiple cancer types. In healthy individuals, CLDN18.2 expression is predominantly limited to gastric mucosa cells and is largely absent from other normal adult tissues . Conversely, CLDN18.2 is highly overexpressed in gastric, gastroesophageal junction, and pancreatic adenocarcinomas, with expression frequencies ranging from 20% to 80% depending on cancer type . This differential expression provides a substantial therapeutic window for targeting cancer cells while minimizing off-target effects on normal tissues.
CLDN18 has two splice variants: CLDN18.1 and CLDN18.2. While CLDN18.1 is specifically expressed in lung tissue, CLDN18.2 has a more restricted expression in normal tissues, detected only in small patches of stomach mucosa . The key challenge in developing CLDN18.2-specific antibodies stems from the high sequence homology between these two isoforms, with only 7-8 amino acid differences in the first extracellular loop . This structural similarity necessitates precision in antibody development to ensure specificity for CLDN18.2 without cross-reactivity to CLDN18.1. Selective binding is essential because targeting the more widely expressed CLDN18.1 could potentially result in unwanted toxicity to normal lung tissue. Therefore, rigorous screening methods, such as high-throughput FACS-based hybridoma screening, are employed to identify antibody clones with exquisite specificity for CLDN18.2 .
The primary method for determining CLDN18.2 expression in clinical and preclinical tumor samples is immunohistochemistry (IHC). Researchers typically use CLDN18.2-specific monoclonal antibodies to perform IHC assays on formalin-fixed, paraffin-embedded tissue sections . This technique allows for visualization of membrane-localized CLDN18.2 expression and assessment of both prevalence and intensity of staining. Through IHC screening, studies have found that approximately 58% of gastric cancers, 60% of gastroesophageal junction cancers, and 20% of pancreatic adenocarcinomas exhibit positive membrane expression of CLDN18.2 . Flow cytometry provides another validation method, particularly useful for cell line studies and for screening antibody clones for selective binding to membrane-bound CLDN18.2 . Additionally, molecular techniques such as RT-PCR or RNA sequencing may be employed to detect CLDN18.2 transcript expression, though protein-level confirmation remains the gold standard for therapeutic targeting purposes.
Developing highly specific CLDN18.2 monoclonal antibodies requires sophisticated immunization and screening strategies. The most successful approaches employ multiplexed immunization protocols, where mice are simultaneously or sequentially exposed to different CLDN18.2 antigen formats . One effective strategy combines: (1) peptide immunization using sequences that span loop 2 of the CLDN18.2 extracellular domain, which contains the isoform-specific amino acids; (2) immunization with cells engineered to overexpress full-length CLDN18.2 protein (such as transfected NIH3T3 cells); and (3) in some cases, DNA immunization . Following immunization, high-throughput FACS-based hybridoma screening is essential for identifying antibody clones with selective binding to membrane-bound CLDN18.2 without cross-reactivity to CLDN18.1 . Lead candidates undergo further validation through binding assays with cells engineered to express either CLDN18.1 or CLDN18.2, along with human cancer cell lines with endogenous expression. Subsequent humanization of promising mouse monoclonal antibodies involves carefully transferring the complementarity-determining regions (CDRs) to human antibody frameworks, followed by affinity maturation if needed to preserve or enhance binding characteristics .
Optimizing ADCC and ADCP mechanisms for CLDN18.2 antibodies involves strategic engineering of the antibody structure and experimental validation across multiple assay systems. For ADCC enhancement, researchers should focus on the Fc region of the antibody, particularly glycoengineering through methods such as afucosylation, which increases binding affinity to FcγRIIIa on NK cells . In vitro ADCC assays typically employ NK cells or PBMCs as effector cells and CLDN18.2-expressing tumor cells as targets, with cytotoxicity measured via release of intracellular markers or metabolic indicators of cell viability.
For ADCP optimization, modifications that enhance binding to FcγRI and FcγRIIa on macrophages and other phagocytic cells are beneficial. In vitro ADCP assays commonly utilize monocyte-derived macrophages or macrophage cell lines as effectors, with phagocytosis quantified through flow cytometry or microscopy-based methods . Isotype selection is critical, with human IgG1 typically providing superior effector function compared to other isotypes. Studies with CLDN18.2 antibodies have demonstrated that Fc effector function is a key component of their mechanism of action, suggesting that optimization of these pathways may significantly enhance therapeutic efficacy .
Developing effective CLDN18.2-directed ADCs requires careful optimization of multiple components. The starting point is selecting a high-affinity, highly selective CLDN18.2 antibody with demonstrated capacity for internalization upon target binding . Internalization efficiency can be measured using fluorescently labeled antibodies and lysosomal tracking markers to confirm that the antibody-receptor complex is efficiently routed to lysosomal compartments after binding .
The linker-payload system requires particular attention. Studies have shown success with cleavable linkers, such as valine-citrulline dipeptide linkers, which enable payload release in the lysosomal environment . For payloads, potent microtubule inhibitors like monomethyl auristatin E (MMAE) have demonstrated efficacy in CLDN18.2-directed ADCs. The drug-to-antibody ratio (DAR) should be optimized to balance potency with potential aggregation or altered pharmacokinetics; typically, a DAR of 2-4 provides a good balance .
In preclinical evaluation, both cell line xenograft (CDX) models and patient-derived xenograft (PDX) models expressing CLDN18.2 should be utilized to assess efficacy . The CLDN18.2-307-ADC, generated by conjugating MMAE to a CLDN18.2 mAb using a cleavable linker, demonstrated remarkable efficacy, inducing complete and sustained tumor regressions in both CDX and PDX models of pancreatic cancer . This comprehensive approach to ADC development maximizes the likelihood of creating a therapeutically viable candidate for clinical evaluation.
When designing animal studies for CLDN18.2 antibodies, researchers must carefully consider species differences in CLDN18.2 expression patterns and antibody cross-reactivity. Most humanized CLDN18.2 antibodies are specifically engineered to recognize human CLDN18.2, which may limit direct testing in immunocompetent mouse models . To address this limitation, researchers should implement a multi-faceted approach:
For initial efficacy studies, immunodeficient mouse models (such as CD-1 nude mice) bearing human tumor xenografts provide valuable insights into direct anti-tumor effects . Both cell line-derived xenografts (CDXs) and patient-derived xenografts (PDXs) with confirmed CLDN18.2 expression should be utilized to capture the heterogeneity of human tumors . Measurement parameters should include not only tumor volume but also molecular analyses of tumor tissue to assess CLDN18.2 modulation, immune infiltration, and pathway alterations.
To evaluate immune-mediated mechanisms like ADCC and ADCP, which are critical for CLDN18.2 antibody efficacy, specialized models are needed. These include using mice reconstituted with human immune cells or developing surrogate antibodies that recognize mouse CLDN18.2 with similar affinity and specificity as the clinical candidate recognizes human CLDN18.2 . Antibody binding to mouse CLDN18.2 should be verified in advance through immunohistochemistry of mouse gastric tissue or other validation methods .
Patient selection for CLDN18.2 antibody clinical trials should be guided by comprehensive biomarker analysis, with CLDN18.2 expression as the primary selection criterion. IHC assessment of CLDN18.2 membrane expression in tumor tissue is essential, with positivity thresholds carefully established based on preclinical efficacy correlations . Screening studies have demonstrated that approximately 58% of gastric cancers, 60% of gastroesophageal junction cancers, and 20% of pancreatic adenocarcinomas exhibit positive membrane expression of CLDN18.2, providing a substantial population for potential treatment .
Beyond expression levels, cellular localization of CLDN18.2 is critical—membrane localization, rather than cytoplasmic expression, correlates with therapeutic accessibility and efficacy . For ADC approaches, internalization capacity may be an additional consideration. Patient selection might also incorporate tumor stage and prior treatment history, with clinical data suggesting that CLDN18.2-positive status may persist in metastatic lesions, providing rationale for including patients with advanced disease .
For combination therapies, additional biomarkers related to immune status or complementary pathway activation may refine patient selection. Importantly, standardized IHC protocols with validated antibodies and scoring systems are essential for consistent patient identification across clinical sites, particularly as CLDN18.2 testing moves toward companion diagnostic development for approved therapies .
Anticipating resistance to CLDN18.2-targeted therapies requires proactive investigation of multiple potential mechanisms. Primary resistance may occur due to heterogeneous CLDN18.2 expression within tumors or rapid internalization and degradation of antibody-target complexes without effective cell killing . To address this, research protocols should include detailed baseline characterization of CLDN18.2 expression patterns across multiple tumor regions and single-cell analysis to quantify the proportion of CLDN18.2-positive cells.
Acquired resistance mechanisms likely include downregulation or mutation of CLDN18.2, alterations in internalization pathways (particularly relevant for ADCs), or immune escape through modulation of effector cell recruitment or function . Serial tumor biopsies or liquid biopsy approaches should be incorporated into research protocols to monitor for these changes during treatment. In preclinical models, generating resistant cell lines through prolonged exposure to CLDN18.2 antibodies can provide valuable insights into adaptation mechanisms.
Combination strategies represent a proactive approach to resistance prevention. These include combining CLDN18.2 antibodies with chemotherapy to enhance tumor cell killing, immune checkpoint inhibitors to potentiate immune-mediated effects, or targeting complementary tumor-associated antigens to address heterogeneous expression . Bispecific antibody formats that simultaneously engage CLDN18.2 and secondary targets represent an emerging approach. Additionally, ADC technologies with novel payloads could overcome resistance mediated by drug efflux or altered intracellular trafficking pathways .
Confirming CLDN18.2 isoform specificity is crucial given the high homology between CLDN18.1 and CLDN18.2. An effective validation strategy employs multiple complementary approaches. The gold standard involves parallel testing on cell lines engineered to express either CLDN18.1 or CLDN18.2 exclusively . Flow cytometry provides quantitative data on differential binding, while Western blotting can confirm specificity at the protein level. Binding kinetics, assessed through surface plasmon resonance or bio-layer interferometry using recombinant CLDN18.1 and CLDN18.2 proteins, provide additional evidence of specificity with quantitative affinity measurements .
Tissue cross-reactivity studies represent another critical validation method. Immunohistochemistry should demonstrate positive staining in CLDN18.2-expressing gastric mucosa while showing minimal reactivity with CLDN18.1-expressing lung tissue . This pattern confirms target selectivity in physiologically relevant contexts. Additionally, epitope mapping through techniques such as hydrogen-deuterium exchange mass spectrometry or X-ray crystallography of antibody-antigen complexes can provide structural confirmation that the antibody binds to regions containing the 7-8 amino acids that differ between the isoforms .
For humanized antibodies, maintaining isoform specificity throughout the humanization process requires careful monitoring. Each humanized variant should undergo the same battery of specificity tests as the original murine antibody to ensure that the critical binding properties are preserved .
Optimizing antibody internalization for CLDN18.2-directed ADCs requires both strategic epitope selection and rigorous experimental validation. The extracellular domain of CLDN18.2 contains two loops that can serve as antibody binding sites, with different epitopes potentially leading to varying internalization efficiencies . Antibodies targeting specific epitopes on the first extracellular loop of CLDN18.2 have demonstrated superior internalization in some studies.
Quantitative assessment of internalization can be performed using confocal microscopy with fluorescently labeled antibodies, allowing visualization of the internalization process and co-localization with endosomal/lysosomal markers . Flow cytometry-based internalization assays, where surface-bound antibody is distinguished from internalized antibody using acid washing or secondary detection reagents, provide quantitative measurement of internalization rates under various conditions . These assays should evaluate internalization across multiple CLDN18.2-positive cell lines to account for potential variations in cellular machinery.
Engineering approaches can enhance internalization efficiency, including modification of antibody binding valency, as bivalent binding can promote receptor clustering and accelerate internalization . Additionally, selecting specific antibody subclasses or engineering the Fc region may influence intracellular trafficking patterns. For the CLDN18.2-307-ADC, research confirmed that upon binding to the extracellular domain, the CLDN18.2-ADC/CLDN18.2 protein complex was efficiently internalized and subsequently localized to the lysosomal compartment, which is essential for releasing the cytotoxic payload .
Comprehensive characterization of CLDN18.2 antibodies requires a multi-parametric analytical approach. Binding affinity and kinetics should be determined through surface plasmon resonance (SPR) or bio-layer interferometry (BLI), providing kon, koff, and KD values that quantify the strength and stability of antibody-antigen interactions . Epitope binning experiments using these same platforms can classify antibodies into groups that recognize overlapping or distinct epitopes.
For functional characterization, in vitro assays measuring various mechanisms of action are essential. ADCC assays using human NK cells or PBMCs as effectors provide insights into immune cell engagement and cytotoxicity . Similarly, ADCP assays using macrophages demonstrate phagocytic potential. For ADCs, cytotoxicity assays across a panel of CLDN18.2-expressing and non-expressing cell lines establish potency and specificity .
Advanced biophysical methods further refine characterization. Hydrogen-deuterium exchange mass spectrometry or X-ray crystallography of antibody-antigen complexes provide atomic-level understanding of binding interfaces . For ADCs, drug-to-antibody ratio (DAR) analysis by mass spectrometry and hydrophobic interaction chromatography ensures consistent conjugation .
In situ methods like immunohistochemistry on tissue microarrays containing multiple tumor and normal tissue types provide critical information about binding specificity in physiologically relevant contexts . This comprehensive analytical package enables researchers to select and optimize antibodies with ideal properties for therapeutic development.
The development of bispecific or multispecific antibodies targeting CLDN18.2 represents a promising frontier in cancer immunotherapy. These engineered molecules could simultaneously engage CLDN18.2 on tumor cells and immune effector cells (such as T cells, NK cells, or macrophages) to enhance anti-tumor immune responses beyond what is achievable with conventional monoclonal antibodies . For T-cell engagement, formats that bind CD3 on T cells and CLDN18.2 on tumor cells could redirect polyclonal T cells to kill CLDN18.2-expressing tumors, potentially overcoming the limitations of conventional antibodies that rely primarily on NK cell-mediated ADCC.
The development process would build upon existing CLDN18.2 antibody discovery platforms, utilizing the highly specific binding domains already identified while incorporating secondary binding domains for immune effectors . Critical considerations include optimizing the affinity of each binding domain to achieve the desired potency while minimizing off-target effects, selecting appropriate linker lengths and flexibility to facilitate effective immune synapse formation, and engineering properties that provide favorable pharmacokinetics and tissue distribution .
Preclinical evaluation would require specialized models that contain both human tumor cells expressing CLDN18.2 and relevant human immune effector populations . Given the restricted expression of CLDN18.2 in normal tissues, this approach could potentially deliver highly potent anti-tumor activity while maintaining an acceptable safety profile, particularly for difficult-to-treat cancers like pancreatic adenocarcinoma where CLDN18.2 expression has been confirmed .
Combining CLDN18.2 antibodies with complementary immunotherapy approaches offers multiple strategies to enhance therapeutic efficacy. One promising approach involves pairing CLDN18.2 antibodies with immune checkpoint inhibitors targeting PD-1/PD-L1 or CTLA-4 pathways . The mechanistic rationale lies in the potential synergy between CLDN18.2 antibody-mediated ADCC, which can initiate immune responses against tumor cells, and checkpoint inhibition, which can prevent subsequent immunosuppression. This combination could be particularly valuable for cancers like gastric and pancreatic adenocarcinomas, where single-agent checkpoint inhibitors have shown limited efficacy.
Another strategic combination involves CLDN18.2 antibodies with immunomodulatory agents that enhance NK cell or macrophage function, such as IL-15 agonists, CD47 inhibitors, or TLR agonists . These combinations could amplify the immune effector mechanisms that underlie CLDN18.2 antibody efficacy. For CLDN18.2-directed ADCs, combinations with agents that modulate the tumor microenvironment to enhance antibody penetration or that target complementary survival pathways could prevent resistance development .
Research protocols investigating these combinations should evaluate not only additive or synergistic anti-tumor effects but also potential enhanced toxicities. Sequential treatment schedules might prove superior to concurrent administration in some cases, necessitating careful optimization of dosing and timing through preclinical modeling before clinical translation .
CLDN18.2 antibodies hold significant potential as diagnostic and theranostic agents, extending their utility beyond direct therapeutic applications. As diagnostic tools, CLDN18.2-specific antibodies are already valuable for immunohistochemical identification of CLDN18.2-positive tumors, serving as companion diagnostics for patient selection in clinical trials . This application could be expanded to develop standardized diagnostic kits with validated scoring systems for consistent assessment across laboratories.
The theranostic potential of CLDN18.2 antibodies lies in their ability to serve dual diagnostic and therapeutic functions when coupled with appropriate imaging agents or radionuclides . Radiolabeled CLDN18.2 antibodies could enable positron emission tomography (PET) or single-photon emission computed tomography (SPECT) imaging to non-invasively identify CLDN18.2-expressing tumors, quantify expression levels, and monitor treatment responses. This approach would be particularly valuable for patient selection and monitoring in trials of CLDN18.2-targeted therapies.
Further innovation could involve developing CLDN18.2 antibody fragments or mimetics with optimized properties for imaging, such as faster clearance and improved tumor penetration. Additionally, antibody-fluorophore conjugates could facilitate intraoperative visualization of CLDN18.2-positive malignancies during surgical procedures. The development of CLDN18.2 theranostics would leverage much of the same binding specificity data generated during therapeutic antibody development, creating opportunities for parallel development of diagnostic and therapeutic applications .