Open Access

Claudin 1, 4, 6 and 18 isoform 2 as targets for the treatment of cancer (Review)

  • Authors:
    • Masuko Katoh
    • Masaru Katoh
  • View Affiliations

  • Published online on: September 13, 2024     https://doi.org/10.3892/ijmm.2024.5424
  • Article Number: 100
  • Copyright: © Katoh et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The 24 claudin (CLDN) genes in the human genome encode 26 representative CLDN family proteins. CLDNs are tetraspan‑transmembrane proteins at tight junctions. Because several CLDN isoforms, such as CLDN6 and CLDN18.2, are specifically upregulated in human cancer, CLDN‑targeting monoclonal antibodies (mAbs), antibody‑drug conjugates (ADCs), bispecific antibodies (bsAbs) and chimeric antigen receptor (CAR) T cells have been developed. In the present review, CLDN1‑, 4‑, 6‑ and 18.2‑targeting investigational drugs in clinical trials are discussed. CLDN18.2‑directed therapy for patients with gastric and other types of cancer is the most advanced area in this field. The mouse/human chimeric anti‑CLDN18.2 mAb zolbetuximab has a single‑agent objective response rate (ORR) of 9%, and increases progression‑free and overall survival in combination with chemotherapy. The human/humanized anti‑CLDN18.2 mAb osemitamab, and ADCs AZD0901, IBI343 and LM‑302, with single‑agent ORRs of 28‑60%, have been tested in phase III clinical trials. In addition, bsAbs, CAR T cells and their derivatives targeting CLDN4, 6 or 18.2 are in phase I and/or II clinical trials. AZD0901, IBI343, zolbetuximab and the anti‑CLDN1 mAb ALE.C04 have been granted fast track designation or priority review designation by the US Food and Drug Administration.

Introduction

The human claudin (CLDN) gene family consists of the CLDN1-12, 14-20, 22-25 and 34 genes (1-4). The CLDN10 gene encodes the CLDN10a and CLDN10b isoforms, expressed under control of renal/uterine and ubiquitous promoters, respectively (5). The CLDN18 gene encodes CLDN18.1 and CLDN18.2 isoforms, expressed under the control of pulmonary and gastric promoters, respectively (6). Therefore, the 24 human CLDN genes encode 26 representative full-length CLDN proteins (Table I).

Table I

Human CLDN gene family.

Table I

Human CLDN gene family.

GeneAliasLocusParacellular functionDisease (genetic alteration)
CLDN1SEMP13q28Paracellular barrierILVASC syndrome (germline mut)
CLDN2-Xq22.3Cation channelOAZON syndrome (germline mut)
CLDN3RVP17q11.23Paracellular barrier-
CLDN4CPETR17q11.23Unclear-
CLDN5TMVCF22q11.21Paracellular barrier-
CLDN6-16p13.3Paracellular barrier-
CLDN7-17p13.1Unclear-
CLDN8-21q22.11Unclear-
CLDN9DFNB11616p13.3Paracellular barrierNon-syndromic deafness (germline mut)
CLDN10a-13q32.1Anion channel (CLDN10a); cation channel (CLDN10b)HELIX syndrome (germline mut)
CLDN11-3q26.2UnclearHLD22 (germline mut)
CLDN12-7q21.13Unclear-
CLDN14DFNB2921q22.13Paracellular barrierNon-syndromic deafness (germline mut)
CLDN15-7q22.1Cation channel-
CLDN16HOMG33q28Cation channelFamilial hypomagnesemia (germline mut)
CLDN17-21q21.3Anion channel-
CLDN18a-3q22.3Paracellular barriersGastric cancer (somatic CLDN18::ARHGAP fus)
CLDN19HOMG51p34.2Cation channelFamilial hypomagnesemia (germline mut)
CLDN20-6q25.3NR-
CLDN22CLDN214q35.1NR-
CLDN23-8p23.1Paracellular barrier-
CLDN24CLDN224q35.1NR-
CLDN25CLDN2411q23.2Cation channel-
CLDN34-Xp22.2NR-

a Two isoforms depending on promoter. ARHGAP, Rho GTPase-activating protein; CLDN, claudin; fus, fusion; HELIX, hypohidrosis, electrolyte imbalance, lacrimal gland dysfunction, ichthyosis and xerostomia; HLD22, hypomyelinating leukodystrophy-22; ILVASC, ichthyosis, leukocyte vacuoles, alopecia and sclerosing cholangitis; mut, mutation; NR, not reported; OAZON, obstructive azoospermia with nephrolithiasis; -, not applicable.

CLDN isoforms at intercellular tight junctions have four transmembrane domains. The first extracellular folding loop stabilizes the paracellular interface and the C-terminal cytoplasmic region interacts with the zona occludens 1 scaffold protein for the assembly of other tight junction proteins (7,8). CLDN proteins that form homo- and heterotypical as well as trans/cis complexes regulate paracellular barrier or permeability functions at endothelial, epidermal, gastrointestinal, renal and other interfaces to maintain organ and/or whole-body homeostasis (9-15). Tight junction functions are dynamically regulated by junctional CLDN isoforms that undergo antegrade transport from the Golgi apparatus to the plasma membrane, endocytosis to early endosomes, and sorting to recycling endosomes for trafficking back to the cell surface or late endosomes for lysosomal degradation (Fig. 1).

Germline mutations in human CLDN genes have been reported in patients with non-cancerous diseases (Table I). CLDN1 mutations are related to ichthyosis, leukocyte vacuoles, alopecia and sclerosing cholangitis syndrome (16). CLDN2 mutations are related to obstructive azoospermia with hypercalciuria and kidney stones syndrome (17). CLDN10 mutations are associated with hypohidrosis, electrolyte imbalance, lacrimal gland dysfunction, ichthyosis and xerostomia syndrome (18). CLDN9 and CLDN14 mutations are associated with non-syndromic deafness (19,20). CLDN16 and CLDN19 mutations are associated with familial hypermagnesemia (21,22). CLDN11 mutations are associated with hypomyelinating leukodystrophy (23).

Somatic CLDN18::Rho GTPase-activating protein (ARHGAP)6, CLDN18::ARHGAP10, CLDN18::ARHGAP26 and CLDN18::ARHGAP42 fusions are detected in 3-15% of gastric cancer cases (24,25). Furthermore, CLDN1 upregulation in head and neck squamous cell carcinoma (HNSCC) and hepatocellular carcinoma (HCC), CLDN4 and CLDN6 upregulation in ovarian and other types of cancer, and CLDN18.2 upregulation in gastric or gastroesophageal junction adenocarcinoma (GEA) and pancreatic ductal adenocarcinoma (PDAC; Fig. 2) have been reported (26-30).

Because CLDN proteins at non-junctional basolateral membranes in tumor cells are accessible targets for antibody-based therapeutic modalities irrespective of their oncogenic or tumor suppressive function and signaling, anti-CLDN monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs) and bispecific antibodies (bsAbs; including bispecific T cell engagers), as well as CLDN-directed chimeric antigen receptor (CAR) T cells, have been developed (29-34). Drugs targeting CLDN1, 4, 6 and 18.2 have entered clinical trials for the treatment of cancer (Table II) and some of them have proceeded to later-phase trials (Fig. 3). Information on CLDN-targeted therapeutics in clinical trials is reviewed subsequently.

Table II

Clinical development of CLDN-targeting therapeutics.

Table II

Clinical development of CLDN-targeting therapeutics.

Target/modalityDrugPhaseClinical trial IDDesignStatus
CLDN1 mAbALE.C04aI/IINCT06054477Mono and comboRecruiting
CLDN4 bsAbASP1002INCT05719558MonoRecruiting
CLDN6 mAbASP1650IINCT03760081MonoCompleted
CLDN6 ADCTORL-1-23INCT05103683MonoRecruiting
CLDN6 bsAbAMG 794INCT05317078MonoActive NR
BNT142I/IINCT05262530MonoRecruiting
XmAb541INCT06276491MonoRecruiting
CLDN6 tsAbSAIL66INCT05735366MonoRecruiting
CLDN6 CARBNT211I/IINCT04503278MonoRecruiting
CAR NK cellsINCT05410717MonoRecruiting
CLDN18.2 mAbAB011INCT04400383Mono and comboCompleted
ASKB589IIINCT06206733ComboRecruiting
FG-M108IIINCT06177041ComboRecruiting
MIL93INCT04671875MonoRecruiting
OsemitamabbIIINCT06093425ComboNyR
ZL-1211I/IINCT05065710MonoCompleted
ZolbetuximabcIIINCT03504397ComboActive NR
CLDN18.2 ADCATG-022bINCT05718895MonoRecruiting
AZD0901aIIINCT06346392MonoRecruiting
EO-3021INCT05980416MonoRecruiting
IBI343aIIINCT06238843MonoRecruiting
LM-302IIINCT06351020MonoRecruiting
SOT102I/IINCT05525286Mono and comboRecruiting
CLDN18.2 bsAbAZD5863I/IINCT06005493MonoRecruiting
GivastomigbINCT04900818MonoRecruiting
GresonitamabINCT04260191MonoTerminated
IBI389INCT05164458Mono and comboRecruiting
PM1032I/IINCT05839106MonoRecruiting
PT886bI/IINCT05482893Mono and comboRecruiting
Q-1802INCT04856150MonoRecruiting
QLS31905I/IINCT06041035ComboNyR
CLDN18.2 CARAZD6422INCT05981235MonoRecruiting
IMC002INCT05472857MonoRecruiting
KD-496INCT05583201MonoRecruiting
LB1908INCT05539430MonoRecruiting
Satri-celI/IINCT04581473MonoRecruiting

a FDA fast track;

b FDA orphan drug;

c FDA priority review, rejected owing to unspecified deficiencies in a third-party manufacturing facility, but resubmitted. ADC, antibody-drug conjugate; bsAb, bispecific antibody; CAR, chimeric antigen receptor; CLDN, claudin; combo, combination therapy; FDA, Food and Drug Administration; mAb, monoclonal antibody; mono, monotherapy; NCT, National Clinical Trial; NK, natural killer; NR, not recruiting; NyR, not yet recruiting; Satri-cel, satricabtagene autoleucel; tsAb, trispecific antibody.

CLDN1-targeted therapy

CLDN1 is transcriptionally upregulated by tumor necrosis factor α signaling to the NF-κB complex, hypoxia signaling to the hypoxia-inducible factor α and β complex and WNT signaling to the β-catenin-T cell factor/lymphoid enhancer factor complex (30,35,36). Because CLDN1 upregulation in HCC maintains cancer stem cells and the protumor immune microenvironment and promotes invasion and metastasis, the prognosis of patients with CLDN1-high HCC is worse than that of patients with CLDN1-low HCC (30). CLDN1 is also upregulated in HNSCC and other types of cancer, such as breast, colorectal, gastric, ovarian, pancreatic and thyroid cancer (37-40).

Anti-CLDN1 mAbs

Humanized anti-CLDN1 mAb derived from a rat anti-human CLDN1 mAb (OM-7D3-B3) exhibits preclinical antitumor activity through the suppression of cancer stemness and tumor invasion and reprogramming of the immunosuppressive tumor microenvironment in HCC xenograft models, especially those with WNT/β-catenin signaling activation and an epithelial-mesenchymal transition phenotype (30,37).

The humanized anti-CLDN1 mAb ALE.C04 has also been shown to have preclinical antitumor activity as a single agent and in combination with immune checkpoint inhibitors in HNSCC xenograft models via perturbation of interactions between tumor and stromal cells to reverse extracellular matrix remodeling, tissue fibrosis and T cell immune evasion (40,41).

ALE.C04 has received fast track designation by the US Food and Drug Administration (FDA) for treatment of recurrent or metastatic CLDN1-positive HNSCC (42). A phase I/II clinical trial of ALE.C04, as a monotherapy and in combination with pembrolizumab, is ongoing, with an estimated completion date of February 2028 [trial no. National Clinical Trial (NCT)06054477; Table II].

CLDN4-targeted therapy

CLDN4 is upregulated in solid tumors, including triple-negative breast cancer (TNBC), colorectal and gastric cancer (intestinal subtype), non-small cell lung cancer (NSCLC), ovarian (serous subtype), pancreatic and prostate cancer, and urothelial carcinoma (26,40,43). The biological roles of CLDN4 in human carcinogenesis are dependent on the tumor type (26,39).

Anti-CLDN4 bsAbs

The bsAb ASP1002, which targets CLDN4 and 4-1BB, has been shown to induce antitumor activity in cancer with CLDN4 upregulation through costimulatory T cell signaling activation and subsequent T cell proliferation and cytokine production (43); however, to the best of our knowledge, single-chain variable fragment (scFv) components targeting CLDN4 and CD137 and their epitopes, in vitro functions and in vivo antitumor effects of ASP1002 are unknown. ASP1002 is in a phase I clinical trial for the treatment of CLDN4-positive solid tumors, such as colorectal, ovarian and prostate cancer, NSCLC, TNBC and urothelial carcinoma (trial no. NCT05719558; Table II).

CLDN6-targeted therapy

CLDN6 is preferentially expressed in pluripotent stem cells and endodermal precursors, such as hepatic or pancreatic progenitor cells, and in fetal tissue derived from the stomach, pancreas and lung, but is suppressed in adult tissues (44-46). CLDN6 is then reactivated in tumor tissue and upregulated in ovarian (14-55%), endometrial (17-21%) and gastric cancer (10-52%), HCC (0-80%), and NSCLC (6-11%), as well as rare malignancies, such as germ cell (54-100%) and atypical teratoid/rhabdoid tumors (29-100%), and myxofibrosarcomas (64%) (28,47-49).

Because CLDN6 is an oncofetal protein or cancer-specific antigen (28,44-49), CLDN6-targeting drugs have been developed for the treatment of cancer. Anti-CLDN6 mAbs (ASP1650) (50-52), ADCs (TORL-1-23) (53,54), CLDN6-targeting bsAbs (AMG 794, BNT142 and XmAb541) (55-57), trispecific antibodies (tsAbs; SAIL66) (58), and CAR T (BNT211) (45,59) and natural killer (NK) cells (60) have been tested in clinical trials (Table II).

Anti-CLDN6 mAbs

ASP1650 is a mouse/human chimeric anti-CLDN6 mAb that exhibited antitumor effects on ovarian cancer and testicular tumor cells via antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity in a preclinical study (50) and has entered a phase I clinical trial for ovarian cancer (trial no. NCT02054351) and phase II clinical trial for male patients with germ cell tumors (trial no. NCT03760081). The objective response rates (ORRs) of ASP1650 were 2 (1/41) and 0% (0/13) in the NCT02054351 and NCT03760081 clinical trials, respectively (51,52).

Anti-CLDN6 ADCs

TORL-1-23 (CLDN6-23-ADC) is an anti-CLDN6 ADC that consists of humanized CLDN6-23-mAb that recognizes the second extracellular loop of CLDN6, a protease cleavable linker and a monomethyl auristatin E (MMAE) cytotoxic payload with a drug/antibody ratio (DAR) of 4.1 (53). In a preclinical study, TORL-1-23 was >10-fold more potent than CLDN6-23-mAb and exerted in vivo antitumor effects on bladder, endometrial and ovarian cancer (53). Notably, in a phase I clinical trial for the treatment of advanced cancer, including endometrial, ovarian and testicular cancer (trial no. NCT05103683), the ORR of TORL-1-23 in CLDN6-positive cancer was 32% (7/22) (54).

Anti-CLDN6 bsAbs

AMG 794, BNT142 and XmAb541 are bsAbs that simultaneously bind CLDN6 on tumor cells and CD3 on T cells for the elimination of tumor cells through recruitment, activation and proliferation of CLDN6-targeting cytotoxic T cells (55-57). Mouse model experiments have revealed that AMG 794 exerted antitumor effects on epithelial ovarian cancer and NSCLC (55) and that BNT142 exerted antitumor effects on serous ovarian cancer and ovarian teratocarcinoma (56). A phase I clinical trial of AMG 794 for patients with NSCLC, epithelial ovarian cancer and other types of solid tumor (trial no. NCT05317078), a phase I/II clinical trial of BNT142 for CLDN6-positive solid tumors (trial no. NCT05262530) and a phase I clinical trial of XmAb541 for solid tumors (trial no. NCT06276491) are ongoing.

Anti-CLDN6 tsAbs

SAIL66 is an anti-CLDN6 tsAb that binds CLDN6 on tumor cells and CD3, as well as 4-1BB on T cells to induce more robust immune reactions in CLDN6-positive tumor cells compared with conventional anti-CLDN6 bsAbs (58). In a preclinical study, SAIL66 successfully enhanced T cell infiltration and mitigated T cell exhaustion via potentiation of 4-1BB-mediated costimulatory signaling in syngeneic mouse model experiments (58). In a phase I clinical trial of SAIL66 for CLDN6-positive solid tumors, the anti-interleukin-6 receptor mAb tocilizumab was added to ameliorate cytokine release syndrome (CRS) caused by enhanced antitumor immunity (trial no. NCT05735366).

CLDN6-directed CAR T cells

BNT211 consists of CLDN6-targeting and 4-1BB-stimulating CAR T cells and a CAR T cell-amplifying RNA vaccine (CARVac) (59). CARVac is a liposomal CLDN6-expressing RNA that induces ectopic CLDN6 expression on antigen-presenting cells, such as dendritic cells and macrophages, for stimulation and expansion of CLDN6-targeting CAR T cells (59). BNT211 exerted antitumor effects on ovarian and lung tumors in a preclinical study (45) and was assessed in a phase I/II clinical trial for the treatment of patients with CLDN6-positive solid tumors (trial no. NCT04503278). Although signs of CRS appeared in ~50% of patients due to enhanced immunity, the ORR of BNT211 was 33% (7/21) (59).

CLDN18.2-targeted therapy

CLDN18.2 is upregulated in gastric (27-56%), esophageal (50%) and ovarian adenocarcinoma (10%), PDAC (30-60%), and NSCLC (4%) (27,61-64). Due to the upregulation of non-junctional CLDN18.2 in tumor tissues and the limited expression of tight junction protein CLDN18.2 in gastric epithelial cells, CLDN18.2 is a quasicancer-specific antigen (34). Currently, >20 drugs directed against CLDN18.2 are in clinical trials for treatment of GEA and other types of cancer (Table II).

Anti-CLDN18.2 mAbs

AB011 (65), ASKB589 (66,67), FG-M108 (68), MIL93 (69), osemitamab (70,71), ZL-1211 (72) and zolbetuximab (73-75) are representative anti-CLDN18.2 mAbs with ADCC activity that have entered clinical trials (Table II). ASKB589, FG-M108 and zolbetuximab have been investigated in phase III clinical trials, and a phase III clinical trial of osemitamab is underway (Fig. 3).

ASKB589, FG-M108 and osemitamab are humanized/human mAbs that had manageable safety profiles and were well-tolerated in early-phase clinical trials (trial nos. NCT04632108/NCT05632939, NCT04894825 and NCT04396821/NCT04495296, respectively). Notably, the single-agent ORR of ASKB589 in solid tumors was 22% (2/9) (66) and that of osemitamab was 58% (23/40) (70). The ORRs of ASKB589 + capecitabine and oxaliplatin (CAPOX) without or with anti-programmed cell death protein 1 (PD-1) mAb sintilimab in GEA were 75 (9/12) and 80% (12/15), respectively (66,67). The ORR of FG-M108 + nab-paclitaxel + gemcitabine in PDAC was 50% (7/14) (68) and the ORR of osemitamab + CAPOX + anti-PD-1 mAb nivolumab was 57% (45/79) in GEA (71). Currently, randomized, double-blind phase III clinical trials of ASKB589 + CAPOX + sintilimab vs. placebo + CAPOX + sintilimab (trial no. NCT06206733) and FG-M108 + CAPOX vs. placebo + CAPOX (trial no. NCT06177041) in first-line settings for CLDN18.2-positive GEA are ongoing. Osemitamab was granted orphan drug designation by the FDA and combination therapy of osemitamab + chemotherapy + nivolumab is advancing toward a phase III clinical trial for the treatment of GEA (trial no. NCT06093425).

Zolbetuximab is a mouse/human chimeric mAb with a single-agent ORR of 9% (4/43) in CLDN18.2-positive GEA (73). The combination of zolbetuximab + modified 5-fluorouracil, leucovorin and oxaliplatin chemotherapy (mFOLFOX6) resulted in greater clinical activity than placebo + mFOLFOX6 in the SPOTLIGHT study (trial no. NCT03504397) for GEA with CLDN18.2 upregulation. The median progression-free survival (mPFS) time was 10.6 vs. 8.7 months [hazard ratio (HR), 0.75; 95% CI, 0.60-0.94] and the median overall survival (mOS) time was 18.2 vs. 15.5 months (HR, 0.75; 95% CI, 0.60-0.94) (74). The combination of zolbetuximab + CAPOX improved clinical activity in comparison with placebo + CAPOX in the GLOW study (trial no. NCT03653507) (75). Zolbetuximab was granted priority review designation by the FDA in July 2023 but rejected due to unspecified deficiencies in a third-party manufacturing facility in January 2024 (76). The data were resubmitted to the FDA in May 2024 and decision is expected in November 2024 under the Prescription Drug User Fee Act (77).

Anti-CLDN18. 2 ADCs

ATG-022 (78), AZD0901 (CMG901) (79,80), EO-3021 (CPO102 or SYSA1801) (81), IBI343 (82), LM-302 (83,84) and SOT102 (85) are representative anti-CLDN18.2 ADCs in clinical trials (Table II). ATG-022, AZD0901, EO-3021 and LM-302 are human/humanized anti-CLDN18.2 mAbs conjugated with MMAE via cleavable linkers, whereas IBI343 and SOT102 are anti-CLDN18.2 ADCs connected to the topoisomerase I inhibitor exatecan and a derivative of PNU-159682 anthracycline, respectively (78-85). AZD0901 has been assessed in a phase III clinical trial, and IBI343 and LM-302 are entering phase III clinical trials (Fig. 3).

AZD0901, with a DAR of 4, has been shown to exert direct cytotoxic effects on CLDN18.2-overexpressing tumor cells and bystander killing effects on surrounding tumor cells in preclinical studies, and had a single-agent ORR of 44% (39/89) in GEA with CLDN18.2 upregulation in the KYM901 phase I clinical trial (trial no. NCT04805307) (79,80). AZD0901 also exhibited manageable safety profiles in the clinic. Anemia (62.8%), vomiting (57.5%) and hypoalbuminemia (57.5%) were common treatment-emergent adverse events (TEAEs), and decreased neutrophil count (18.6%) and anemia (13.3%) were the most frequent grade ≥3 TEAEs (79). AZD0901 monotherapy for treatment of CLDN18.2-overexpressing GEA was granted fast track designation by the FDA in April 2022 (86) and is currently in a phase II clinical trial with an expected completion date of May 2025 (trial no. NCT06219941); another phase III randomized clinical trial of AZD0901 monotherapy vs. apatinib, docetaxel, irinotecan, paclitaxel or TAS-102, has an expected completion date of April 2026 (trial no. NCT06346392).

IBI343 had single-agent ORRs of 28 [7/25 patients with PDAC and biliary tract cancer (BTC)] and 40% (4/10 patients with CLDN18.2-positive PDAC) in a phase I clinical trial (trial no. NCT05458219) and a manageable safety profile with any-grade [anemia (37%), nausea (26%), vomiting (26%) and decreased white blood cell count (20%)] and grade ≥3 treatment-related adverse events (TRAEs) [anemia (6%) and decreased white blood cell counts (3%)] (82). IBI343 monotherapy was granted fast track designation by the FDA for the treatment of PDAC (87). By contrast, for the treatment of GEA, a phase II clinical trial of combination therapy of IBI343 + sintilimab (trial no. NCT06321913) and a phase III randomized clinical trial of IBI343 vs. irinotecan or paclitaxel, (trial no. NCT06238843) are planned but not yet recruiting.

LM-302 had superior antitumor efficacy to zolbetuximab in a preclinical study using gastric cancer model (83), and a single-agent ORR of 31% (11/36) in CLDN18.2-positive GEA and a manageable safety profile in a phase I/II clinical trial (trial no. NCT05161390) (84). LM-302 is proceeding to a phase III randomized clinical trial of LM-302 vs. the investigator's choice of therapy, apatinib or irinotecan, for treatment of CLDN18.2-positive GEA (trial no. NCT06351020).

Anti-CLDN18.2 bsAbs

AZD5863 (88), givastomig (89), gresonitamab (90), IBI389 (91), PM1032 (92,93), PT886 (94), Q-1802 (95) and QLS31905 (96) are representative anti-CLDN18.2 bsAbs (Table II).

AZD5863, gresonitamab, IBI389 and QLS31905 simultaneously binds CLDN18.2 on tumor cells and CD3 on T cells (88,90,91,96), which activates CD3 signaling in T cells and redirects cytotoxic T cells toward tumor killing through lytic synapse formation and release of granzymes and perforins (97,98).

Givastomig, PM1032, PT886 and Q-1802 simultaneously bind CLDN18.2 and non-CD3 immune antigens; givastomig and PM1032 enhance antitumor immunity via 4-1BB-induced activation of costimulatory signaling (89,92); PT886 reactivates phagocytosis via CD47-dependent inhibition of 'do not eat me' signaling (94); and Q-1802 inhibits immune checkpoints via programmed death-ligand 1 (PD-L1)/PD-1 signaling blockade (95). Givastomig and PT886 have been granted orphan drug designation by the FDA (94,99).

Compared with anti-CLDN18.2 mAbs and ADCs, anti-CLDN18.2 bsAbs are in relatively early phases of clinical trials (88-96): Givastomig, IBI389 and Q-1802 are in phase I clinical trials; AZD5863, PM1032 and PT886 are in phase I/II clinical trials; and QLS31905 is proceeding to phase I/II clinical trials (Fig. 3).

In a phase I clinical trial, IBI389 caused any-grade TRAEs in 97.4% (111/114) and grade ≥3 TRAEs in 55.3%, including any-grade CRS (57.0%) and grade 3 CRS (0.9%), of patients with solid tumors, such as GEA (n=37) and PDAC (n=66) (trial no. NCT05164458). The single-agent ORR of IBI389 was 31% (8/26) in GEA with CLDN18.2 overexpression (91).

In a phase I/II clinical trial, PM1032 caused any-grade TRAEs in 73% (22/30) and grade ≥3 TRAEs in 10% of gastrointestinal cancer cases (trial no. NCT05839106). The single-agent ORR of PM1032 was 20% (2/10) in CLDN18.2-positive GEA (93).

In a phase I clinical trial (trial no. NCT04856150) of Q-1802, any-grade TRAEs, including nausea (62%; 18/29), vomiting (62%) and abdominal pain (28%), immune-related adverse events (AEs; such as abnormal thyroid function, rash and arthritis) (24%), and grade 3 TRAEs such as nausea and vomiting (24%) and grade 4 TRAE hyponatremia (3%) were observed. The single-agent ORR of Q-1802 was 22% (2/9) in gastrointestinal cancer (95).

QLS31905 treatment was associated with any-grade TRAEs in 98% (51/52) and grade ≥3 TRAEs in 40% of patients treated with 0.5-500.0 mg/kg once/week (qW) or once every 2 weeks (q2W) QLS31905, including grade 3 CRS in 2 patients in the 350 mg/kg qW QLS31905 cohort, in a phase I clinical trial (trial no. NCT05278832). The single-agent ORR of QLS31905 was 11% (3/27) in the phase I study (96), and a phase I/II clinical trial of QLS31905 plus chemotherapy is planned (trial no. NCT06041035).

CLDN18.2-directed CAR-T cells

Satricabtagene autoleucel (Satri-cel) refers to autologous CAR T cells targeting CLDN18.2: CAR T cells were generated from peripheral blood mononuclear cells via lentiviral transduction of a second-generation CAR construct consisting of an extracellular humanized anti-CLDN18.2 scFv and a CD8α hinge region, CD28 transmembrane domain and cytoplasmic CD28 costimulatory and CD3ζ signaling domains. CAR T cells were formulated and infused back following preconditioning combination therapy with cyclophosphamide, fludarabine and nab-paclitaxel or gemcitabine (100,101).

Satri-cel was assessed in a phase I clinical trial (trial no. NCT03874897) for the treatment of solid tumors on the basis of the results of a preclinical study that revealed antitumor effects with persistent infiltration of CAR T cells into CLDN18.2-positive gastric cancer patient-derived xenograft models (100). A total of ~75% of patients in the Satri-cel clinical trial received bridging therapy, such as folinic acid, fluorouracil and irinotecan, nab-paclitaxel or irinotecan, during autologous CAR T cell production (median, 27 days; range, 22-187 days) (101). Satri-cel exhibited tolerability and safety profiles with manageable AEs, such as preconditioning-associated transient hematological toxicity (grade ≥3, 100%), CRS (any grade, 97%; grade ≥3, 0%), nausea (any grade, 67%; grade ≥3, 1%), vomiting (any grade, 53%; grade ≥3, 3%) and gastric mucosal injury (grade 1/2, 7%; grade 3, 1%) and without immune effector cell-associated neurotoxicity syndrome, and clinical activity, as indicated by an ORR of 39% (38/98), mPFS time of 4.4 months (95% CI, 3.7-6.6) and mOS time of 8.8 months (95% CI, 7.1-10.2) (101). Currently, a randomized phase I/II clinical trial of Satri-cel vs. apatinib, irinotecan or paclitaxel, for the treatment of CLDN18.2-positive GEA and PDAC is ongoing (trial no. NCT04581473).

CLDN18.2-targeting CAR T cells, such as LB1908 expressing a CAR with a 4-1BB costimulatory domain (102), IMC002 harboring a CAR with anti-CLDN18.2 variable heavy domain of heavy chain antibody instead of a scFv (103), AZD6422-armed CAR T cells with a dominant-negative TGF-β type II receptor (dnTGFBR2) to overcome the TGF-β-induced immunosuppressive tumor microenvironment (104) and KD-496 bispecific CAR T cells that simultaneously recognize CLDN18.2 and NK group 2 member D (NKG2D) ligands on tumor cells (105), are also in phase I clinical trials for the treatment of patients with CLDN18.2-positive tumors (Table II).

Future research directions

CLDN-directed therapy poses issues related to antibody-based therapeutic modalities. Perspectives on mAbs, ADCs, bsAbs and CAR drugs are discussed subsequently, with a focus on CLDN targeting for cancer treatment.

mAbs

Phase III SPOTLIGHT and GLOW clinical trials for CLDN18.2-positive GEA demonstrated greater clinical activity of zolbetuximab + chemotherapy than placebo + chemotherapy (74,75). Other anti-CLDN18.2 mAbs have been assessed, and ASKB589, FG-M108 and osemitamab are being assessed in randomized phase III clinical trials with expected completion dates of December 2026, January 2027 and October 2025, respectively (Fig. 3). The single-agent ORR of chimeric mAb zolbetuximab is 9% (73), whereas that of humanized mAbs ASKB589 and osemitamab, which have increased affinity and enhanced ADCC effects, is 22 and 58%, respectively (66,70). Previous-generation mAb drugs were chimeric antibodies that elicit host immune responses to remaining variable regions derived from mouse antibody, while current-generation mAb drugs are human/humanized antibodies without mouse-derived variable regions (106). Comparison of the clinical activity (mPFS and mOS) of zolbetuximab and other humanized/human anti-CLDN18.2 mAbs is warranted.

ADCs

Anti-CLDN ADCs (78-85) are classified into two types. Most ADCs (CLDN6, TORL-1-23; CLDN18.2, ATG-022, AZD0901, EO-3021 and LM-302) include a human/humanized mAb, cleavable linker and an MMAE microtubule inhibitor. Others have cytotoxic DNA-damaging payloads (CLDN18.2, IBI343 and SOT102; Table II). In general, the clinical activity of these anti-CLDN ADCs is affected by mAb epitope affinity and modification, linker chemistry, and payload pharmacology (107,108).

CLDN-targeting ADCs have exhibited improved clinical outcomes over CLDN-targeting chimeric mAbs in early-phase clinical trials; the single-agent ORRs of ATG-022 (78), AZD0901 (79), EO-3021 (81), IBI343 (82), LM-302 (84) and TORL-1-23 (54) were 20, 44, 38, 28, 31 and 32%, respectively, compared with 0-9% for the chimeric anti-CLDN mAbs ASP1650 (51,52) and zolbetuximab (73).

ORRs of the aforementioned anti-CLDN ADCs are similar to those of FDA-approved ADCs targeting other tumor-associated antigens, such as the ORR of 31.5% of anti-trophoblast cell surface antigen 2 ADC sacituzumab govitecan (109) and the ORR of 42.9% of the anti-nectin cell adhesion molecule 4 ADC enfortumab vedotin (110). AZD0901, IBI343 and LM-302 have entered or are entering phase III clinical trials, which will yield mPFS and mOS data in the future.

bsAbs

Anti-CLDN bsAbs in phase I or I/II clinical trials (Table II) are classified into CD3-directed (CLDN6-targeting AMG 794, BNT142 and XmAb541; and CLDN18.2-targeting AZD5863, gresonitamab, IBI389 and QLS31905) and non-CD3-directed (CLDN4-targeting ASP1002; and CLDN18.2-targeting givastomig, PM1032, PT886 and Q-1802) (43,55-57,88-96). IBI389, PM1032, Q-1802 and QLS31905 have single-agent ORRs of 31, 20, 22 and 11%, respectively (91,93,95,96). A study on gresonitamab was terminated due to business decisions of Amgen (trial no. NCT04260191).

In clinical oncology, bsAbs such as amivantamab (targeting EGFR and MET) (111), petosemtamab (targeting EGFR and leucine-rich repeat-containing G-protein coupled receptor 5) (112) and zenocutuzumab (targeting human EGFR 2 and 3) (113) dually target tumor-specific antigens (97,98). Since upregulation of CLDN18.2 (56%) (27), EGFR (27%) (114), HER2 (21%) (115), fibroblast growth factor receptor 2 isoform b (4-5%) (116,117) and MET (24%) (118) in gastric cancer has been detected and mAbs targeting these receptor tyrosine kinases (RTKs) have been developed (111,112,119), bsAbs dually targeting CLDN18.2 and RTKs may serve as antibody-based drugs for the treatment of gastric cancer in the future.

CAR T or CAR-NK cells

Satri-cel, an autologous CAR T cell therapy with a second-generation CLDN18.2-directed CAR, has an ORR of 39% (101), which is similar to the ORRs of BNT211 (CLDN6-targeting CAR-T cell + RNA vaccine; 33%) (59) and other solid tumor-targeting CAR T cell therapies directed at carcinoembryonic antigen (15%; 6/40) (120), EGFR (18%; 2/11) (121), glypican 3 (50%; 11/22) (122) and guanylyl cyclase 2C (26%; 5/19) (123), but less effective than FDA-approved CAR T cell therapies targeting hematological malignancies, such as the CD19-directed CAR T cells axicabtagene ciloleucel (82%; 83/101) (124) and B cell maturation antigen-directed CAR T cells ciltacabtagene autoleucel (98%; 95/97) (125).

Numerous challenges hinder the activities of CAR T cells, especially those in solid tumors. These include epitope-losing subclonal replacement on the basis of pretreatment heterogeneity and/or posttreatment evolution (126-128), decreased infiltration of CAR T cells into an immunosuppressive tumor microenvironment harboring angiogenic endothelial cells, extracellular matrix-remodeling cancer-associated fibroblasts, regulatory T cells, M2-type macrophages and monocytic myeloid-derived suppressor cells (129-131), and decreased persistence of CAR T cells due to mechanisms similar to the exhaustion of effector T cells with diminished anti-tumor activity dependent on TGF-β signaling as well as PD-L1/PD-1 and other coinhibitory signaling (132-134).

bsCAR T cells dually targeting CLDN18.2 and NKG2D ligands have been developed to address intratumor heterogeneity (105). CLDN6-directed CAR-NK cells, which incorporate the NKG2D transmembrane domain, CD244 costimulatory domain and the DNAX-activation protein 10 signaling domain have been developed to enhance antitumor immunity (60). CLDN18.2-directed CAR T cells armed with dnTGFBR2 have been developed to eliminate immunosuppressive effects of TGF-β (104) and preclinical CAR T cells armed with forkhead-box O1 transcription factor have been developed to maintain the stem/memory cell subpopulation, retain effector functions and prevent the exhaustion phenotype (133,134). These strategies might further improve the clinical activity of CLDN-targeted CAR therapy.

Conclusion

Drugs directed at CLDN1, 4, 6 and 18.2 tumor-associated antigens have been developed on the basis of mAb, ADC, bsAb or CAR modalities. A total of >30 CLDN-targeting therapeutics have entered clinical trials, and anti-CLDN18.2 mAbs (ASKB589, FG-M108, osemitamab and zolbetuximab) and ADCs (AZD0901, IBI343 and LM-302) have been assessed in phase III clinical trials. AZD0901, IBI343, zolbetuximab and anti-CLDN1 mAb ALE.C04 have been granted fast track or priority review designation by the FDA but have not yet been approved as of August 2024. However, there is a lack of results from phase III clinical trials other than those involving zolbetuximab. Monotherapies with human/humanized ADCs and armed CAR T cells may be promising choices for CLDN-directed cancer therapy.

Availability of data and materials

Not applicable.

Authors' contributions

MasukoK and MasaruK wrote the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Katoh Fund for Knowledge-Base and the Global Network Projects.

References

1 

Tsukita S, Furuse M and Itoh M: Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol. 2:285–293. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Katoh M and Katoh M: CLDN23 gene, frequently down-regulated in intestinal-type gastric cancer, is a novel member of CLAUDIN gene family. Int J Mol Med. 11:683–689. 2003.PubMed/NCBI

3 

Krause G, Winkler L, Mueller SL, Haseloff RF, Piontek J and Blasig IE: Structure and function of claudins. Biochim Biophys Acta. 1778:631–645. 2008. View Article : Google Scholar

4 

Baltzegar DA, Reading BJ, Brune ES and Borski RJ: Phylogenetic revision of the claudin gene family. Mar Genomics. 11:17–26. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Günzel D, Stuiver M, Kausalya PJ, Haisch L, Krug SM, Rosenthal R, Meij IC, Hunziker W, Fromm M and Müller D: Claudin-10 exists in six alternatively spliced isoforms that exhibit distinct localization and function. J Cell Sci. 122:1507–1517. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Niimi T, Nagashima K, Ward JM, Minoo P, Zimonjic DB, Popescu NC and Kimura S: Claudin-18, a novel downstream target gene for the T/EBP/NKX2.1 homeodomain transcription factor, encodes lung- and stomach-specific isoforms through alternative splicing. Mol Cell Biol. 21:7380–7390. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Zihni C, Mills C, Matter K and Balda MS: Tight junctions: From simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Biol. 17:564–580. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Vecchio AJ, Rathnayake SS and Stroud RM: Structural basis for Clostridium perfringens enterotoxin targeting of claudins at tight junctions in mammalian gut. Proc Natl Acad Sci USA. 118:e20246511182021. View Article : Google Scholar : PubMed/NCBI

9 

Günzel D and Yu AS: Claudins and the modulation of tight junction permeability. Physiol Rev. 93:525–569. 2013. View Article : Google Scholar : PubMed/NCBI

10 

France MM and Turner JR: The mucosal barrier at a glance. J Cell Sci. 130:307–314. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Stamatovic SM, Johnson AM, Sladojevic N, Keep RF and Andjelkovic AV: Endocytosis of tight junction proteins and the regulation of degradation and recycling. Ann N Y Acad Sci. 1397:54–65. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Horowitz A, Chanez-Paredes SD, Haest X and Turner JR: Paracellular permeability and tight junction regulation in gut health and disease. Nat Rev Gastroenterol Hepatol. 20:417–432. 2023. View Article : Google Scholar : PubMed/NCBI

13 

Meoli L and Günzel D: The role of claudins in homeostasis. Nat Rev Nephrol. 19:587–603. 2023. View Article : Google Scholar : PubMed/NCBI

14 

Tanaka H, Yamamoto Y, Kashihara H, Yamazaki Y, Tani K, Fujiyoshi Y, Mineta K, Takeuchi K, Tamura A and Tsukita S: Claudin-21 has a paracellular channel role at tight junctions. Mol Cell Biol. 36:954–964. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Raya-Sandino A, Lozada-Soto KM, Rajagopal N, Garcia-Hernandez V, Luissint AC, Brazil JC, Cui G, Koval M, Parkos CA, Nangia S and Nusrat A: Claudin-23 reshapes epithelial tight junction architecture to regulate barrier function. Nat Commun. 14:62142023. View Article : Google Scholar : PubMed/NCBI

16 

Hadj-Rabia S, Baala L, Vabres P, Hamel-Teillac D, Jacquemin E, Fabre M, Lyonnet S, De Prost Y, Munnich A, Hadchouel M and Smahi A: Claudin-1 gene mutations in neonatal sclerosing cholangitis associated with ichthyosis: A tight junction disease. Gastroenterology. 127:1386–1390. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Askari M, Karamzadeh R, Ansari-Pour N, Karimi-Jafari MH, Almadani N, Sadighi Gilani MA, Gourabi H, Vosough Taghi Dizaj A, Mohseni Meybodi A, Sadeghi M, et al: Identification of a missense variant in CLDN2 in obstructive azoospermia. J Hum Genet. 64:1023–1032. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Klar J, Piontek J, Milatz S, Tariq M, Jameel M, Breiderhoff T, Schuster J, Fatima A, Asif M, Sher M, et al: Altered paracellular cation permeability due to a rare CLDN10B variant causes anhidrosis and kidney damage. PLoS Genet. 13:e10068972017. View Article : Google Scholar : PubMed/NCBI

19 

Sineni CJ, Yildirim-Baylan M, Guo S, Camarena V, Wang G, Tokgoz-Yilmaz S, Duman D, Bademci G and Tekin M: A truncating CLDN9 variant is associated with autosomal recessive nonsyndromic hearing loss. Hum Genet. 138:1071–1075. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Wilcox ER, Burton QL, Naz S, Riazuddin S, Smith TN, Ploplis B, Belyatseva I, Ben-Yosef T, Liburd NA, Morell RJ, et al: Mutations in the gene encoding tight junction claudin-14 cause autosomal recessive deafness DFNB29. Cell. 104:165–172. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Simon DB, Lu Y, Choate KA, Velazquez H, Al-Sabban E, Praga M, Casari G, Bettinelli A, Colussi G, Rodriguez-Soriano J, et al: Paracellin-1, a renal tight junction protein required for paracellular Mg2+ resorption. Science. 285:103–106. 1999. View Article : Google Scholar : PubMed/NCBI

22 

Konrad M, Schaller A, Seelow D, Pandey AV, Waldegger S, Lesslauer A, Vitzthum H, Suzuki Y, Luk JM, Becker C, et al: Mutations in the tight-junction gene claudin 19 (CLDN19) are associated with renal magnesium wasting, renal failure, and severe ocular involvement. Am J Hum Genet. 79:949–957. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Riedhammer KM, Stockler S, Ploski R, Wenzel M, Adis-Dutschmann B, Ahting U, Alhaddad B, Blaschek A, Haack TB, Kopajtich R, et al: De novo stop-loss variants in CLDN11 cause hypomyelinating leukodystrophy. Brain. 144:411–419. 2021. View Article : Google Scholar :

24 

Cancer Genome Atlas Research Network: Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 513:202–209. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Nakayama I, Shinozaki E, Sakata S, Yamamoto N, Fujisaki J, Muramatsu Y, Hirota T, Takeuchi K, Takahashi S, Yamaguchi K and Noda T: Enrichment of CLDN18-ARHGAP fusion gene in gastric cancers in young adults. Cancer Sci. 110:1352–1363. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Morin PJ: Claudin proteins in human cancer: Promising new targets for diagnosis and therapy. Cancer Res. 65:9603–9606. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Sahin U, Koslowski M, Dhaene K, Usener D, Brandenburg G, Seitz G, Huber C and Türeci O: Claudin-18 splice variant 2 is a pan-cancer target suitable for therapeutic antibody development. Clin Cancer Res. 14:7624–7634. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Turksen K and Troy TC: Junctions gone bad: Claudins and loss of the barrier in cancer. Biochim Biophys Acta. 1816:73–79. 2011.PubMed/NCBI

29 

Qu H, Jin Q and Quan C: CLDN6: From traditional barrier function to emerging roles in cancers. Int J Mol Sci. 22:134162021. View Article : Google Scholar : PubMed/NCBI

30 

Roehlen N, Muller M, Nehme Z, Crouchet E, Jühling F, Del Zompo F, Cherradi S, Duong FHT, Almeida N, Saviano A, et al: Treatment of HCC with claudin-1-specific antibodies suppresses carcinogenic signaling and reprograms the tumor microenvironment. J Hepatol. 78:343–355. 2023. View Article : Google Scholar

31 

Katoh M and Katoh M: Precision medicine for human cancers with Notch signaling dysregulation (Review). Int J Mol Med. 45:279–297. 2020.PubMed/NCBI

32 

Cao W, Xing H, Li Y, Tian W, Song Y, Jiang Z and Yu J: Claudin18.2 is a novel molecular biomarker for tumor-targeted immunotherapy. Biomark Res. 10:382022. View Article : Google Scholar : PubMed/NCBI

33 

Vonniessen B, Tabariès S and Siegel PM: Antibody-mediated targeting of Claudins in cancer. Front Oncol. 14:13207662024. View Article : Google Scholar : PubMed/NCBI

34 

Nakayama I, Qi C, Chen Y, Nakamura Y, Shen L and Shitara K: Claudin 18.2 as a novel therapeutic target. Nat Rev Clin Oncol. 21:354–369. 2024. View Article : Google Scholar : PubMed/NCBI

35 

Miwa N, Furuse M, Tsukita S, Niikawa N, Nakamura Y and Furukawa Y: Involvement of claudin-1 in the beta-catenin/Tcf signaling pathway and its frequent upregulation in human colorectal cancers. Oncol Res. 12:469–476. 2001. View Article : Google Scholar

36 

Katoh M: Multi-layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β-catenin signaling activation (Review). Int J Mol Med. 42:713–725. 2018.PubMed/NCBI

37 

Zeisel MB, Dhawan P and Baumert TF: Tight junction proteins in gastrointestinal and liver disease. Gut. 68:547–561. 2019. View Article : Google Scholar

38 

Bhat AA, Syed N, Therachiyil L, Nisar S, Hashem S, Macha MA, Yadav SK, Krishnankutty R, Muralitharan S, Al-Naemi H, et al: Claudin-1, a double-edged sword in cancer. Int J Mol Sci. 21:5692020. View Article : Google Scholar : PubMed/NCBI

39 

Hana C, Thaw Dar NN, Galo Venegas M and Vulfovich M: Claudins in cancer: A current and future therapeutic target. Int J Mol Sci. 25:46342024. View Article : Google Scholar : PubMed/NCBI

40 

Toso A, Teixiera G, Zimmermann T, Schmitter D, Meyer M, Muller M, Mailly L, Baumert T and Iacone R: 193P CLAUDIN-1 targeting antibodies in solid tumors: From ALE.C04 to CLAUDIN-1 oncology platform. Immunooncol Technol. 16(Suppl 1): S1003052022. View Article : Google Scholar

41 

Toso A, Teixeira G, Zimmermann T, Gill SG, Schmitter D, Meyer M, Muller M, Mailly L, Baumert T, Manenti L and Iacone R: Abstract LB284: CLAUDIN-1 targeting antibody ALE.C04 drives single activity and restores anti-PD1 efficacy in solid tumors. Cancer Res. 83(Suppl 8): LB2842023. View Article : Google Scholar

42 

Rosa K: FDA grantsfast track status to ALE.C04 for recurrent or metastatic CLDN1+ HNSCC. OncLive. 2023, https://www.onclive.com/view/fda-grants-fast-track-status-to-ale-c04-for-recurrent-or-metastatic-cldn1-hnscc.

43 

Pelster M, Marron TU, Friend BD, Fan A, Yang J and Spira AI: Phase 1 study of ASP1002, a bispecific antibody targeting claudin 4 (CLDN4) and CD137, in patients with locally advanced (LA) or metastatic solid tumors that express CLDN4. J Clin Oncol. 42(Suppl 16): TPS26702024. View Article : Google Scholar

44 

Ben-David U, Nudel N and Benvenisty N: Immunologic and chemical targeting of the tight-junction protein Claudin-6 eliminates tumorigenic human pluripotent stem cells. Nat Commun. 4:19922013. View Article : Google Scholar : PubMed/NCBI

45 

Reinhard K, Rengstl B, Oehm P, Michel K, Billmeier A, Hayduk N, Klein O, Kuna K, Ouchan Y, Wöll S, et al: An RNA vaccine drives expansion and efficacy of claudin-CAR-T cells against solid tumors. Science. 367:446–453. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Kong FE, Li GM, Tang YQ, Xi SY, Loong JHC, Li MM, Li HL, Cheng W, Zhu WJ, Mo JQ, et al: Targeting tumor lineage plasticity in hepatocellular carcinoma using an anti-CLDN6 antibody-drug conjugate. Sci Transl Med. 13:eabb62822021. View Article : Google Scholar : PubMed/NCBI

47 

Zhang C, Guo C, Li Y, Liu K, Zhao Q and Ouyang L: Identification of claudin-6 as a molecular biomarker in pan-cancer through multiple omics integrative analysis. Front Cell Dev Biol. 9:7266562021. View Article : Google Scholar : PubMed/NCBI

48 

Du H, Yang X, Fan J and Du X: Claudin 6: Therapeutic prospects for tumours, and mechanisms of expression and regulation (Review). Mol Med Rep. 24:6772021. View Article : Google Scholar : PubMed/NCBI

49 

Tsang N, Veillard N, Horsley E, Havenith K, Janghra N, Zeitseva K, Oblette C, Kirby I, Hogg PW, Zammarchi F, et al: Abstract 3122: Preclinical development of a novel camptothecin-based antibody-drug conjugate targeting solid tumors expressing Claudin-6. Cancer Res. 84(76 Suppl): S31222024. View Article : Google Scholar

50 

Türeci Ö, Kreuzberg M, Walter K, Wöll S, Schmitt R, Mitnacht-Kraus R, Nakajo I, Yamada T and Sahin U: Abstract 882: The anti-claudin 6 antibody, IMAB027, induces antibody-dependent cellular and complement-dependent cytotoxicity in claudin 6-expressing cancer cells. Cancer Res. 78(Suppl 13): S8822018. View Article : Google Scholar

51 

Sahin U, Jaeger D, Marme F, Mavratzas A, Krauss J, De Greve J, Vergote I and Tureci O: First-in-human phase I/II dose-escalation study of IMAB027 in patients with recurrent advanced ovarian cancer (OVAR): Preliminary data of phase I part. J Clin Oncol. 33(15 Suppl): S55372015. View Article : Google Scholar

52 

Adra N, Vaughn DJ, Einhorn LH, Hanna NH, Funt SA, Rosales M, Arozullah A and Feldman DR: A phase II study assessing the safety and efficacy of ASP1650 in male patients with relapsed refractory germ cell tumors. Invest New Drugs. 40:1087–1094. 2022. View Article : Google Scholar : PubMed/NCBI

53 

McDermott MSJ, O'Brien NA, Hoffstrom B, Gong K, Lu M, Zhang J, Luo T, Liang M, Jia W, Hong JJ, et al: Preclinical efficacy of the antibody-drug conjugate CLDN6-23-ADC for the treatment of CLDN6-positive solid tumors. Clin Cancer Res. 29:2131–2143. 2023. View Article : Google Scholar : PubMed/NCBI

54 

Konecny GE, Wahner Hendrickson AE, Winterhoff B, Machado A, Chander C, Davenport S, Bilic S, Miller LL, Chung A, Press MF, et al: 756P First-in-human phase I study of a novel claudin 6 (CLDN6) targeted antibody drug conjugate (ADC) TORL-1-23. Ann Oncol. 34(Suppl 2): S5172023. View Article : Google Scholar

55 

Pham E, Henn A, Sable B, Wahl J, Conner K, Matthes K, Gupta S, Yabut R, Aeffner F, Wilson KL, et al: Abstract 5202: AMG 794, a Claudin 6-targeted half-life extended (HLE) bispecific T cell engager (BITE®) molecule for non-small cell lung cancer and epithelial ovarian cancer. Cancer Res. 82(Suppl 12): S52022022. View Article : Google Scholar

56 

Stadler CR, Ellinghaus U, Fischer L, Bähr-Mahmud H, Rao M, Lindemann C, Chaturvedi A, Scharf C, Biermann I, Hebich B, et al: Preclinical efficacy and pharmacokinetics of an RNA-encoded T cell-engaging bispecific antibody targeting human claudin 6. Sci Transl Med. 16:eadl27202024. View Article : Google Scholar : PubMed/NCBI

57 

Faber MS, Lee SH, Kim YK, Qi J, Avery KN, Nguyen DHT, Rashid R, Eivazi A, Chu SY, Diaz JE, et al: Abstract 1860: Bispecific claudin-6 x CD3 antibodies in a 2 + 1 format demonstrate selectivity and activity on human ovarian cancer cells. Cancer Res. 81(Suppl 13): S18602021. View Article : Google Scholar

58 

Kamikawa T, Kimura N, Ishii S, Muraoka M, Taniguchi K, Uchikawa R, Yoshimoto M, Okuda-Miura M, Akai S, Kodama T, et al: 1172 SAIL66, a next generation of T cell engager targeting CLDN6, potentiates efficacy. J Immunother Cancer. 11(Suppl 1): S11722023.

59 

Mackensen A, Haanen JBAG, Koenecke C, Alsdorf W, Wagner-Drouet E, Borchmann P, Heudobler D, Ferstl B, Klobuch S, Bokemeyer C, et al: CLDN6-specific CAR-T cells plus amplifying RNA vaccine in relapsed or refractory solid tumors: The phase 1 BNT211-01 trial. Nat Med. 29:2844–2853. 2023. View Article : Google Scholar : PubMed/NCBI

60 

Li J, Hu H, Lian H, Yang S, Liu M, He J, Cao B, Chen D, Hu Y, Zhi C, et al: NK-92MI cells engineered with anti-claudin-6 chimeric antigen receptors in immunotherapy for ovarian cancer. Int J Biol Sci. 20:1578–1601. 2024. View Article : Google Scholar : PubMed/NCBI

61 

Micke P, Mattsson JSM, Edlund K, Lohr M, Jirström K, Berglund A, Botling J, Rahnenfuehrer J, Marincevic M, Pontén F, et al: Aberrantly activated claudin 6 and 18.2 as potential therapy targets in non-small-cell lung cancer. Int J Cancer. 135:2206–2214. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Dottermusch M, Krüger S, Behrens HM, Halske C and Röcken C: Expression of the potential therapeutic target claudin-18.2 is frequently decreased in gastric cancer: Results from a large Caucasian cohort study. Virchows Arch. 475:563–571. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Chen J, Xu Z, Hu C, Zhang S, Zi M, Yuan L and Cheng X: Targeting CLDN18.2 in cancers of the gastrointestinal tract: New drugs and new indications. Front Oncol. 13:11323192023. View Article : Google Scholar : PubMed/NCBI

64 

Lyu SI, Fretter C, Simon AG, Spielmann SM, Damanakis AI, Zhao Y, Bruns CJ, Schmidt T, Popp FC, Waldschmidt D, et al: Extent and clinical significance of the therapy-relevant tight junction protein Claudin 18.2 in pancreatic ductal adenocarcinoma-real-world evidence. Transl Oncol. 47:1020442024. View Article : Google Scholar

65 

Li J, Pan H, Liu T, Xu N, Zhang Y, Qin Y, Shi J, Liao D, Shen L, Luo S, et al: A multicenter, phase 1 study of AB011, a recombinant humanized anti-CLDN18.2 monoclonal antibody, as monotherapy and combined with capecitabine and oxaliplatin (CAPOX) in patients with advanced solid tumors. J Clin Oncol. 41(Suppl 4): S3912023. View Article : Google Scholar

66 

Zhang M, Gong J, Wang J, Shi J, Zhu H, Wang Y, Chen Y, Wang F, Qu X, Yu J, et al: A phase I/II study of ASKB589 [anti-claudin 18.2 (CLDN18.2) monoclonal antibody] in patients with solid tumors. J Clin Oncol. 41(Suppl 4): S3972023. View Article : Google Scholar

67 

Peng Z, Shen L, He Y, Chen J, Hickingbottom B and Lu J: A phase Ib/II study of ASKB589 [anti-Claudin 18.2 (CLDN18.2) monoclonal antibody] combined with CAPOX and PD-1 inhibitor as first-line treatment for locally advanced, relapsed and metastatic gastric/gastro-esophageal junction (G/GEJ) adenocarcinoma. J Clin Oncol. 42(Suppl 3): S3172024. View Article : Google Scholar

68 

Jin Z, Zhang Y, Liu F, Zhang S, Gong J, Zhang M, Liang X, Wang J, Li Y, Yang X, et al: FG-M108 plus nab-paclitaxel and gemcitabine (AG) as first-line (1L) treatment for patients with Claudin-18.2 (CLDN18.2) positive locally advanced unresectable or metastatic pancreatic cancer (PC): Preliminary results from the phase 1b study. J Clin Oncol. 42(Suppl 16): S41422024. View Article : Google Scholar

69 

Huang J, Zhang B, Wang Y, Wang F, Yu Z, Wu S, Zheng Y, Cao Y, Xu J, Lan D, et al: Safety and preliminary efficacy of MIL93 in patients with advanced solid tumors: The monotherapy part of a phase 1 trial. J Clin Oncol. 41(Suppl 4): S7982023. View Article : Google Scholar

70 

Janjigian Y, Tolcher A, Mehta R, Cecchini M, Van Tine B, Kundranda M, Olatunji A, Patel MR, Berlin J, Rocha-Lima CMSP, et al: Abstract CT132: A Phase I/IIa clinical trial (TranStar101) to evaluate the safety, tolerability and pharmacokinetics of OSEMITAMAB administered as monotherapy or in combination with nivolumab or standard of care in patients with locally advanced or metastatic solid tumors. Cancer Res. 84(Suppl 7): CT1322024. View Article : Google Scholar

71 

Zhang X, Guo Z, Zhang J, Guo W, Sun M, Xu N, Qi C, Zhu X, Zhang L, Qian X, et al: First-line osemitamab (TST001) plus nivolumab and capox for advanced g/GEJ cancer (TranStar102): Results of cohort G from a phase I/IIa study. J Clin Oncol. 42(Suppl 16): S40482024. View Article : Google Scholar

72 

Sharma S, Starodub A, Xu N, Chaudhry A, Sun M, Pelster M, Fu Y, Zhang X, Huang Z, Liu W and Hsu K: Preliminary results of a phase 1/2, first-in-human, open-label, dose escalation study of ZL-1211 (anti-Claudin 18.2 mAb) in patients with unresectable or metastatic solid tumors. J Clin Oncol. 41(Suppl 16): S25372023. View Article : Google Scholar

73 

Türeci O, Sahin U, Schulze-Bergkamen H, Zvirbule Z, Lordick F, Koeberle D, Thuss-Patience P, Ettrich T, Arnold D, Bassermann F, et al: A multicentre, phase IIa study of zolbetuximab as a single agent in patients with recurrent or refractory advanced adenocarcinoma of the stomach or lower oesophagus: The MONO study. Ann Oncol. 30:1487–1495. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Shitara K, Lordick F, Bang YJ, Enzinger P, Ilson D, Shah MA, Van Cutsem E, Xu RH, Aprile G, Xu J, et al: Zolbetuximab plus mFOLFOX6 in patients with CLDN18.2-positive, HER2-negative, untreated, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma (SPOTLIGHT): A multicentre, randomised, double-blind, phase 3 trial. Lancet. 401:1655–1668. 2023. View Article : Google Scholar : PubMed/NCBI

75 

Shah MA, Shitara K, Ajani JA, Bang YJ, Enzinger P, Ilson D, Lordick F, Van Cutsem E, Gallego Plazas J, Huang J, et al: Zolbetuximab plus CAPOX in CLDN18.2-positive gastric or gastroesophageal junction adenocarcinoma: The randomized, phase 3 GLOW trial. Nat Med. 29:2133–2141. 2023. View Article : Google Scholar : PubMed/NCBI

76 

Ra J: FDA denies approval for Astellas' investigational gastric cancer drug. Pharmaceutical Technology. 2014, https://www.pharmaceutical-technology.com/news/fda-denies-approval-for-astellas-investigational-gastric-cancer-drug/.

77 

Conroy R: FDA acknowledges zolbetuximab BLA resubmission for CLDN18.2+ gastric cancer. CancerNetwork. 2024, https://www.cancernetwork.com/view/fda-acknowledges-zolb-etuximab-bla-resubmission-for-cldn18-2-gastric-cancer/.

78 

Bishnoi S, Cao D, Mendis SR, Coward J, Zhao J, Xie H and Zheng L: An open-label, multicenter, phase I study of ATG-022 in patients with advanced/metastatic solid tumors (CLINCH). J Clin Oncol. 42(Suppl 16): S30322024. View Article : Google Scholar

79 

Xu RH, Ruan DY, Zhang DS, Liu FR, Luo SX, Zhuang ZX, Wang ZN, Liu FN, Zhang YQ, Yang JW, et al: A phase 1 trial of claudin 18.2-specific antibody-drug conjugate CMG901 in patients with advanced gastric/gastroesophageal junction cancer. J Clin Oncol. 41(Suppl 36): S4344202023. View Article : Google Scholar

80 

Raufi AG, Goyal L, Smyth E, Szekeres P, Petrone M, Hobson R, Thress K, Origuchi M, Nehra J, Brown JS, et al: CLARITY-PanTumor01: A phase 2 trial of the claudin 18.2-specific antibody-drug conjugate AZD0901 (CMG901) in patients with CLDN18.2-expressing advanced solid tumors. J Clin Oncol. 42(Suppl 16): TPS31632024. View Article : Google Scholar

81 

Wang Y, Gong J, Lin R, Zhao S, Wang J, Wang Q, Zhang Y, Su D, Zhang J, Dong Q, et al: First-in-human dose escalation and expansion study of SYSA1801, an antibody-drug conjugate targeting claudin 18.2 in patients with resistant/refractory solid tumors. J Clin Oncol. 41(Suppl 16): S30162023. View Article : Google Scholar

82 

Yu X, Zhang J, Tazbirkova A, Yang J, Yue J, Sun Y, Pan Y, Sun M, Qin Y, Shen L, et al: Safety and efficacy of IBI343 (anti-claudin18.2 antibody-drug conjugate) in patients with advanced pancreatic ductal adenocarcinoma or biliary tract cancer: Preliminary results from a phase 1 study. J Clin Oncol. 42(Suppl 16): S30372024. View Article : Google Scholar

83 

Huang W, Li Y, Liu Z, Rodon L, Correia S, Li Y and Li R: Preclinical activity for TPX-4589 (LM-302), an antibody-drug conjugate targeting tight junction protein CLDN18.2 in solid tumors. Eur J Cancer. 174(Suppl 1): S41–S42. 2022. View Article : Google Scholar

84 

Bai C, Xue J, Zheng Y, Sun M, Ying J, Zhou F, Yu Y, Sun Y, Xing L, Zhang Y, et al: A phase 1/2 study of LM-302, an anti-claudin 18.2 (CLDN18.2) antibody-drug conjugate in patients with advanced gastric/gastroesophageal junction cancer. J Clin Oncol. 42(Suppl 16): S30282024. View Article : Google Scholar

85 

Spisek R: 2P SOT102, a novel CLDN18.2-targeting antibody-drug conjugate for gastric and pancreatic cancer with a wide range of the tumor target expression. ESMO Open. 8(1 Suppl 2): S1011962023. View Article : Google Scholar

86 

Rosa K: CMG901 elicits responses in CLDN18.2-expressing gastric/GEJ cancer. OncLive. 2023, https://www.onclive.com/view/cmg901-elicits-responses-in-cldn18-2-expressing-gastric-gej-cancer/.

87 

Wahner A: IBI343 receives FDA fast track designation for advanced/metastatic PDAC. OncLive. 2024, https://www.onclive.com/view/ibi343-receives-fda-fast-track-designation-for-advanced-metastatic-pdac/.

88 

Gaspar M, Natoli M, Castan L, Rahmy S, Kelton C, Mulgrew K, Korade M, Huhn O, Rees DG, Sigurdardottir A, et al: 1169 AZD5863: A specific, potent, affinity-optimized claudin 18.2 and CD3 binding T cell-engager that elicits low cytokine release and is capable of bystander killing. J Immunother Cancer. 11(Suppl 1): S11692023.

89 

Gao J, Wang Z, Jiang W, Zhang Y, Meng Z, Niu Y, Sheng Z, Chen C, Liu X, Chen X, et al: CLDN18.2 and 4-1BB bispecific antibody givastomig exerts antitumor activity through CLDN18.2-expressing tumor-directed T-cell activation. J Immunother Cancer. 11:e0067042023. View Article : Google Scholar : PubMed/NCBI

90 

Xu Y, Fu J, Henderson M, Lee F, Jurcak N, Henn A, Wahl J, Shao Y, Wang J, Lyman M, et al: CLDN18.2 BiTE engages effector and regulatory T cells for antitumor immune response in preclinical models of pancreatic cancer. Gastroenterology. 165:1219–1232. 2023. View Article : Google Scholar : PubMed/NCBI

91 

Zheng L, Ruihong D, Jieer Y, Xu Q, Guo Z, Hu C, Sun Y, Niu Z, Hao J, Zhang M, et al: Safety and preliminary efficacy results of IBI389, an anti-CLDN18.2/CD3 bispecific antibody, in patients with solid tumors and gastric or gastro-esophageal tumors: A phase 1 dose escalation and expansion study. J Clin Oncol. 42(Suppl 16): S25192024. View Article : Google Scholar

92 

Wang J, Dong T, Gong X, Li D, Sun J, Luo Y and Wu H: Safety and pharmacokinetic assessment of the FIC CLDN18.2/4-1BB bispecific antibody in rhesus monkeys. Int J Toxicol. 43:291–300. 2024. View Article : Google Scholar

93 

Guo Y, Wu L, Li Y, Wen J, Xue J, Wang Z, Li P, Zhao W, Liu J, Rao X, et al: First-in-human phase I/II safety and preliminary efficacy of PM1032, a bispecific antibody targeting CLDN18.2 and 4-1BB, in patients with advanced solid tumors. J Clin Oncol. 42(Suppl 16): S26622024. View Article : Google Scholar

94 

Overman MJ, Melhem R, Blum-Murphy MA, Ramos C, Petrosyan L, Li J, Perer JK, Zou H, Wang M and Wright HM: A phase I, first-in-human, open-label, dose escalation and expansion study of PT886 in adult patients with advanced gastric, gastroesophageal junction, and pancreatic adenocarcinomas. J Clin Oncol. 41(Suppl 4): TPS7652023. View Article : Google Scholar

95 

Yk W, Gong J, Sun Y, Zhang J, Ni S, Hou J, Chen X, Wang Y, Yu Q, Qu X, et al: Interim results of a first-in-human phase 1 study of Q-1802, a CLDN18.2/PD-L1 bsABs, in patients with relapsed or refractory solid tumors. J Clin Oncol. 41(Suppl 4): S3822023. View Article : Google Scholar

96 

Wang Y, Gong J, Sun Y, Yang S, Zhang M, Cui J, Lv J, Su H, Wang J, Lu J, et al: 132P A phase I clinical trial of QLS31905 in advanced solid tumors. Immunooncol Technol. 20(Suppl): S1006042023. View Article : Google Scholar

97 

Klein C, Brinkmann U, Reichert JM and Kontermann RE: The present and future of bispecific antibodies for cancer therapy. Nat Rev Drug Discov. 23:301–319. 2024. View Article : Google Scholar : PubMed/NCBI

98 

Goebeler ME, Stuhler G and Bargou R: Bispecific and multispecific antibodies in oncology: Opportunities and challenges. Nat Rev Clin Oncol. 21:539–560. 2024. View Article : Google Scholar : PubMed/NCBI

99 

Tucker N: FDA grants orphan drug designation to TJ-CD4B for gastric cancer. Targeted Oncology. 2022, https://www.targetedonc.com/view/fda-grants-orphan-drug-designation-to-tj-cd4b-for-gastric-cancer/.

100 

Jiang H, Shi Z, Wang P, Wang C, Yang L, Du G, Zhang H, Shi B, Jia J, Li Q, et al: Claudin18.2-specific chimeric antigen receptor engineered T cells for the treatment of gastric cancer. J Natl Cancer Inst. 111:409–418. 2019. View Article : Google Scholar

101 

Qi C, Liu C, Gong J, Liu D, Wang X, Zhang P, Qin Y, Ge S, Zhang M, Peng Z, et al: Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial final results. Nat Med. 30:2224–2234. 2024. View Article : Google Scholar : PubMed/NCBI

102 

Zhen DB, Thota R, del Corral C, Geng D, Yang T, Wang C, Amato G, Akram M, Miller DS, Bubuteishvili-Pacaud L and Gibson M: A phase 1, open-label, dose escalation and expansion, multicenter study of claudin 18.2-targeted chimeric antigen receptor T-cells in patients with unresectable, locally advanced, or metastatic gastric, gastroesophageal junction, esophageal, or pancreatic adenocarcinoma. J Clin Oncol. 41(Suppl 4): TSP4802023. View Article : Google Scholar

103 

Luo T, Lu Z, Zheng R, Zhou J, Wang S, Hao R and Sun M: Outstanding safety and efficacy data of IMC002, an autologous CLDN18.2-targeting CAR-T, in CLDN18.2+ advanced solid tumors. J Clin Oncol. 42(Suppl 16): e160122024. View Article : Google Scholar

104 

Britton Z, Breen S, Carrasco R, Clark B, Broggi MAS, Lapointe JM, Giraldo N, Rao Attili BMN, Hatke A, Grigoriadou C, et al: 235 Preclinical evaluation and anti-tumor activity of AZD6422, a CLDN18.2 targeting armored CAR-T for gastric, esophageal and pancreatic cancers. J Immunother Cancer. 11(Suppl 1): A1–A1731. 2023.

105 

Xu H, Li W, Lv H, Gu D, Wei X and Dai H: Tandem CAR-T cells targeting CLDN18.2 and NKG2DL for treatment of gastric cancer. J Clin Oncol. 40(Suppl 16): S40302022. View Article : Google Scholar

106 

Paul S, Konig MF, Pardoll DM, Bettegowda C, Papadopoulos N, Wright KM, Gabelli SB, Ho M, van Elsas A and Zhou S: Cancer therapy with antibodies. Nat Rev Cancer. 24:399–426. 2024. View Article : Google Scholar : PubMed/NCBI

107 

Fu Z, Li S, Han S, Shi C and Zhang Y: Antibody drug conjugate: the 'biological missile' for targeted cancer therapy. Signal Transduct Target Ther. 7:932022. View Article : Google Scholar

108 

Fuentes-Antrás J, Genta S, Vijenthira A and Siu LL: Antibody-drug conjugates: In search of partners of choice. Trends Cancer. 9:339–354. 2023. View Article : Google Scholar : PubMed/NCBI

109 

Kalinsky K, Diamond JR, Vahdat LT, Tolaney SM, Juric D, O'Shaughnessy J, Moroose RL, Mayer IA, Abramson VG, Goldenberg DM, et al: Sacituzumab govitecan in previously treated hormone receptor-positive/HER2-negative metastatic breast cancer: Final results from a phase I/II, single-arm, basket trial. Ann Oncol. 31:1709–1718. 2020. View Article : Google Scholar : PubMed/NCBI

110 

Rosenberg J, Sridhar SS, Zhang J, Smith D, Ruether D, Flaig TW, Baranda J, Lang J, Plimack ER, Sangha R, et al: EV-101: A phase I study of single-agent enfortumab vedotin in patients with nectin-4-positive solid tumors, including metastatic urothelial carcinoma. J Clin Oncol. 38:1041–1049. 2020. View Article : Google Scholar : PubMed/NCBI

111 

Park K, Haura EB, Leighl NB, Mitchell P, Shu CA, Girard N, Viteri S, Han JY, Kim SW, Lee CK, et al: Amivantamab in EGFR exon 20 insertion-mutated non-small-cell lung cancer progressing on platinum chemotherapy: Initial results from the CHRYSALIS phase I study. J Clin Oncol. 39:3391–3402. 2021. View Article : Google Scholar : PubMed/NCBI

112 

Fayette J, Clatot F, Brana I, Saada E, van Herpen CML, Mazard T, Perez CA, Tabernero J, Le Tourneau C, Hollebecque A, et al: Petosemtamab (MCLA-158) with pembrolizumab as first-line (1L) treatment of recurrent/metastatic (r/m) head and neck squamous cell carcinoma (HNSCC): Phase 2 study. J Clin Oncol. 42(Suppl 16): S60142024. View Article : Google Scholar

113 

Schram AM, Goto K, Kim DW, Martin-Romano P, Ou SHI, O'Kane GM, O'Reilly EM, Umemoto K, Duruisseaux M, Neuzillet C, et al: Efficacy and safety of zenocutuzumab, a HER2 x HER3 bispecific antibody, across advanced NRG1 fusion (NRG1+) cancers. J Clin Oncol. 40(Suppl 16): S1052022. View Article : Google Scholar

114 

Kim MA, Lee HS, Lee HE, Jeon YK, Yang HK and Kim WH: EGFR in gastric carcinomas: Prognostic significance of protein overexpression and high gene copy number. Histopathology. 52:738–746. 2008. View Article : Google Scholar : PubMed/NCBI

115 

Van Cutsem E, Bang YJ, Feng-Yi F, Xu JM, Lee KW, Jiao SC, Chong JL, López-Sanchez RI, Price T, Gladkov O, et al: HER2 screening data from ToGA: Targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer. 18:476–484. 2015. View Article : Google Scholar :

116 

Ahn S, Lee J, Hong M, Kim ST, Park SH, Choi MG, Lee JH, Sohn TS, Bae JM, Kim S, et al: FGFR2 in gastric cancer: Protein overexpression predicts gene amplification and high H-index predicts poor survival. Mod Pathol. 29:1095–1103. 2016. View Article : Google Scholar : PubMed/NCBI

117 

Yashiro M, Kuroda K, Masuda G, Okuno T, Miki Y, Yamamoto Y, Sera T, Sugimoto A, Kushiyama S, Nishimura S, et al: Clinical difference between fibroblast growth factor receptor 2 subclass, type IIIb and type IIIc, in gastric cancer. Sci Rep. 11:46982021. View Article : Google Scholar : PubMed/NCBI

118 

Lee HE, Kim MA, Lee HS, Jung EJ, Yang HK, Lee BL, Bang YJ and Kim WH: MET in gastric carcinomas: Comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer. 107:325–333. 2012. View Article : Google Scholar : PubMed/NCBI

119 

Katoh M, Loriot Y, Brandi G, Tavolari S, Wainberg ZA and Katoh M: FGFR-targeted therapeutics: Clinical activity, mechanisms of resistance and new directions. Nat Rev Clin Oncol. 21:312–329. 2024. View Article : Google Scholar : PubMed/NCBI

120 

Zhang H, Yang Z, Zhu X, Li J, Gao Y, Zhang Y, Tong Z, Fu Q, Bao X, Li B, et al: Phase I trial of hypoxia-responsive CEA CAR-T cell therapy in patients with heavily pretreated solid tumor via intraperitoneal or intravenous transfusion. J Clin Oncol. 42(Suppl 16): S35142024. View Article : Google Scholar

121 

Feng K, Guo Y, Dai H, Wang Y, Li X, Jia H and Han W: Chimeric antigen receptor-modified T cells for the immunotherapy of patients with EGFR-expressing advanced relapsed/refractory non-small cell lung cancer. Sci China Life Sci. 59:468–479. 2016. View Article : Google Scholar : PubMed/NCBI

122 

Zhang Q, Fu Q, Cao W, Wang H, Xu X, Huang J, Zou A, Zhu J, Wan H, Yao Y, et al: Phase I study of C-CAR031, a GPC3-specific TGFβRIIDN armored autologous CAR-T, in patients with advanced hepatocellular carcinoma (HCC). J Clin Oncol. 42(Suppl 16): S40192024. View Article : Google Scholar

123 

Qi C, Liu C, Li J, Gong J, Wang X, Wang Z, Lu X, He T, Ding Y, Wu F, et al: Phase I study of GUCY2C CAR-T therapy IM96 in patients with metastatic colorectal cancer. J Clin Oncol. 42(Suppl 16): S25182024. View Article : Google Scholar

124 

Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, Braunschweig I, Oluwole OO, Siddiqi T, Lin Y, et al: Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 377:2531–2544. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Martin T, Usmani SZ, Berdeja JG, Agha M, Cohen AD, Hari P, Avigan D, Deol A, Htut M, Lesokhin A, et al: Ciltacabtagene autoleucel, an anti-B-cell maturation antigen chimeric antigen receptor T-cell therapy, for relapsed/refractory multiple myeloma: CARTITUDE-1 2-year follow-up. J Clin Oncol. 41:1265–1274. 2023. View Article : Google Scholar

126 

Majzner RG and Mackall CL: Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 8:1219–1226. 2018. View Article : Google Scholar : PubMed/NCBI

127 

Shah NN and Fry TJ: Mechanisms of resistance to CAR T cell therapy. Nat Rev Clin Oncol. 16:372–385. 2019.PubMed/NCBI

128 

Larson RC and Maus MV: Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat Rev Cancer. 21:145–161. 2021. View Article : Google Scholar : PubMed/NCBI

129 

Hou AJ, Chen LC and Chen YY: Navigating CAR-T cells through the solid-tumour microenvironment. Nat Rev Drug Discov. 20:531–550. 2021. View Article : Google Scholar : PubMed/NCBI

130 

Tauriello DVF, Sancho E and Batlle E: Overcoming TGFβ-mediated immune evasion in cancer. Nat Rev Cancer. 22:25–44. 2022. View Article : Google Scholar

131 

Katoh M and Katoh M: WNT signaling and cancer stemness. Essays Biochem. 66:319–331. 2022. View Article : Google Scholar : PubMed/NCBI

132 

Gumber D and Wang LD: Improving CAR-T immunotherapy: Overcoming the challenges of T cell exhaustion. EBioMedicine. 77:1039412022. View Article : Google Scholar : PubMed/NCBI

133 

Chan JD, Scheffler CM, Munoz I, Sek K, Lee JN, Huang YK, Yap KM, Saw NYL, Li J, Chen AXY, et al: FOXO1 enhances CAR T cell stemness, metabolic fitness and efficacy. Nature. 629:201–210. 2024. View Article : Google Scholar : PubMed/NCBI

134 

Doan AE, Mueller KP, Chen AY, Rouin GT, Chen Y, Daniel B, Lattin J, Markovska M, Mozarsky B, Arias-Umana J, et al: FOXO1 is a master regulator of memory programming in CAR T cells. Nature. 629:211–218. 2024. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2024
Volume 54 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Katoh M and Katoh M: Claudin 1, 4, 6 and 18 isoform 2 as targets for the treatment of cancer (Review). Int J Mol Med 54: 100, 2024.
APA
Katoh, M., & Katoh, M. (2024). Claudin 1, 4, 6 and 18 isoform 2 as targets for the treatment of cancer (Review). International Journal of Molecular Medicine, 54, 100. https://doi.org/10.3892/ijmm.2024.5424
MLA
Katoh, M., Katoh, M."Claudin 1, 4, 6 and 18 isoform 2 as targets for the treatment of cancer (Review)". International Journal of Molecular Medicine 54.5 (2024): 100.
Chicago
Katoh, M., Katoh, M."Claudin 1, 4, 6 and 18 isoform 2 as targets for the treatment of cancer (Review)". International Journal of Molecular Medicine 54, no. 5 (2024): 100. https://doi.org/10.3892/ijmm.2024.5424