Open Access

Precision medicine for human cancers with Notch signaling dysregulation (Review)

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

  • Published online on: December 4, 2019     https://doi.org/10.3892/ijmm.2019.4418
  • Pages: 279-297
  • 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

NOTCH1, NOTCH2, NOTCH3 and NOTCH4 are transmembrane receptors that transduce juxtacrine signals of the delta‑like canonical Notch ligand (DLL)1, DLL3, DLL4, jagged canonical Notch ligand (JAG)1 and JAG2. Canonical Notch signaling activates the transcription of BMI1 proto‑oncogene polycomb ring finger, cyclin D1, CD44, cyclin dependent kinase inhibitor 1A, hes family bHLH transcription factor 1, hes related family bHLH transcription factor with YRPW motif 1, MYC, NOTCH3, RE1 silencing transcription factor and transcription factor 7 in a cellular context‑dependent manner, while non‑canonical Notch signaling activates NF‑κB and Rac family small GTPase 1. Notch signaling is aberrantly activated in breast cancer, non‑small‑cell lung cancer and hematological malignancies, such as T‑cell acute lymphoblastic leukemia and diffuse large B‑cell lymphoma. However, Notch signaling is inactivated in small‑cell lung cancer and squamous cell carcinomas. Loss‑of‑function NOTCH1 mutations are early events during esophageal tumorigenesis, whereas gain‑of‑function NOTCH1 mutations are late events during T‑cell leukemogenesis and B‑cell lymphomagenesis. Notch signaling cascades crosstalk with fibroblast growth factor and WNT signaling cascades in the tumor microenvironment to maintain cancer stem cells and remodel the tumor microenvironment. The Notch signaling network exerts oncogenic and tumor‑suppressive effects in a cancer stage‑ or (sub)type‑dependent manner. Small‑molecule γ‑secretase inhibitors (AL101, MRK‑560, nirogacestat and others) and antibody‑based biologics targeting Notch ligands or receptors [ABT‑165, AMG 119, rovalpituzumab tesirine (Rova‑T) and others] have been developed as investigational drugs. The DLL3‑targeting antibody‑drug conjugate (ADC) Rova‑T, and DLL3‑targeting chimeric antigen receptor‑modified T cells (CAR‑Ts), AMG 119, are promising anti‑cancer therapeutics, as are other ADCs or CAR‑Ts targeting tumor necrosis factor receptor superfamily member 17, CD19, CD22, CD30, CD79B, CD205, Claudin 18.2, fibroblast growth factor receptor (FGFR)2, FGFR3, receptor‑type tyrosine‑protein kinase FLT3, HER2, hepatocyte growth factor receptor, NECTIN4, inactive tyrosine‑protein kinase 7, inactive tyrosine‑protein kinase transmembrane receptor ROR1 and tumor‑associated calcium signal transducer 2. ADCs and CAR‑Ts could alter the therapeutic framework for refractory cancers, especially diffuse‑type gastric cancer, ovarian cancer and pancreatic cancer with peritoneal dissemination. Phase III clinical trials of Rova‑T for patients with small‑cell lung cancer and a phase III clinical trial of nirogacestat for patients with desmoid tumors are ongoing. Integration of human intelligence, cognitive computing and explainable artificial intelligence is necessary to construct a Notch‑related knowledge‑base and optimize Notch‑targeted therapy for patients with cancer.

1. Introduction

NOTCH1, NOTCH2, NOTCH3 and NOTCH4 are cell surface receptors that transduce juxtacrine signals of delta-like canonical Notch ligand (DLL)1, DLL3, DLL4, jagged canonical Notch ligand (JAG)1 and JAG2 from adjacent cells (1-3). Germline mutations in the NOTCH1, NOTCH2 and NOTCH3 genes cause Adams-Oliver syndrome, Alagille syndrome and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, respectively (4), and DLL4-NOTCH3 signaling in human vascular organoids induces basement membrane thickening and drives vasculopathy in the diabetic microenvironment (5). By contrast, somatic alterations in the genes encoding Notch signaling components drive various types of human cancer, such as breast cancer, small-cell lung cancer (SCLC) and T-cell acute lymphoblastic leukemia (T-ALL) (6-9). Notch signaling dysregulation is involved in a variety of pathologies, including cancer and non-cancerous diseases.

Small-molecule inhibitors, antagonistic monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), bispecific antibodies or biologics (bsAbs) and chimeric antigen receptor-modified T cells (CAR-Ts) targeting Notch signaling components have been developed as investigational anti-cancer drugs (10-12). The safety, tolerability and anti-tumor effects of these compounds have been studied in clinical trials; however, Notch-targeted therapeutics are not yet approved for the treatment of patients with cancer. Here, Notch signaling in the tumor microenvironment and Notch-targeted therapeutics are reviewed, and perspectives on Notch-related precision oncology are discussed with emphases on biologics, clinical sequencing and explainable artificial intelligence.

2. Notch signaling overview

DLL1, DLL3, DLL4, JAG1 and JAG2 are transmembrane ligands of Notch receptors (2,6,13). DLL1, DLL4, JAG1 and JAG2 are agonistic Notch ligands (Fig. 1), whereas DLL3 without the conserved N-terminal module of agonistic Notch ligands is an aberrant Notch ligand that can antagonize DLL1-Notch signaling. EGF-like repeats 1-13 in the extracellular region of NOTCH1 are involved in DLL1/4 signaling and the EGF-like repeats 10-24 of NOTCH1 are involved in JAG1/2 signaling (14). β-1,3-N-Acetylglucosaminyltransferase lunatic fringe and β-1,3-N-acetylglucosaminyltransferase manic fringe transfer N-acetylglucosamine to O-fucose on the EGF repeats in the extracellular region of Notch receptors, which enhances DLL1-NOTCH1 signaling and inhibits JAG1-NOTCH1 signaling (15). DLL1 promotes myogenesis through transient NOTCH1 activation, whereas DLL4 inhibits myogenesis through sustained NOTCH1 activation (16). The expression profile of DLL/JAG ligands and extracellular modification of Notch receptors affect receptor-ligand interactions and modulate the outputs and strength of the Notch signaling cascades (17); however, the landscape of interactions between Notch ligands and receptors, especially those of NOTCH2, NOTCH3 and NOTCH4, remain elusive.

Figure 1

Overview of canonical and non-canonical Notch signaling cascades. DLL/JAG agonistic ligands trigger proteolytic cleavage of Notch receptors to generate the NECD, NTMD and NICD. Canonical Notch signaling cascades: NICD/CSL-dependent transcriptional activation of target genes, such as BMI1, CCND1, CD44, HES1, HEY1, MYC, NOTCH3, REST and TCF7, in a cellular context-dependent manner. Non-canonical Notch signaling cascades: CSL-independent cellular responses, such as NTMD-dependent activation of RAC1, NICD-dependent activation of NF-κB and NICD-dependent inhibition of ATM. DLL4-NOTCH1 signaling in endothelial cells induces NTMD-mediated assembly of cadherin-5, receptor-type tyrosine-protein phosphatase F and TRIO and F-actin-binding protein, which activates RAC1 to maintain vascular barrier function through cytoskeletal reorganization. By contrast, NOTCH1 activation in T-cell acute lymphoblastic leukemia leads to the interaction of NICD with the IκB kinase complex and ATM to activate NF-κB-dependent transcription and inhibit ATM-dependent DNA-damage response, respectively. DLL, delta-like canonical Notch ligand; JAG, jagged canonical Notch ligand; NECD, Notch extracellular domain; NTMD, Notch transmembrane domain; NICD, Notch intracellular domain; ADAM10, disintegrin and metalloproteinase domain-containing protein 10; ATM, serine-protein kinase ATM; MAML, mastermind like protein; CSL, CBF1-suppressor of hairless-LAG1; BMI1, BMI1 proto-oncogene polycomb ring finger; CCND1, cyclin D1; HES1, hes family bHLH transcription factor 1; HEY1, hes related family bHLH transcription factor with YRPW motif 1; REST, RE1 silencing transcription factor; TCF7, transcription factor 7; RAC1, Ras-related protein Rac1.

Interactions with DLL/JAG agonistic ligands trigger sequential proteolytic cleavage of Notch receptors by disintegrin and metalloproteinase domain-containing protein (ADAM)10/17 and γ-secretase (2,6,18,19), which generates the following: i) Notch extracellular domain; ii) Notch transmembrane domain (NTMD); and iii) Notch intracellular domain (NICD) (Fig. 1). The NICD is then translocated to the nucleus and associates with CBF1-suppressor of hairless-LAG1 (CSL) and mastermind like proteins (MAML1, MAML2 or MAML3) to activate the transcription of target genes. NICD/CSL-dependent transcription of Notch target genes is defined as the canonical Notch signaling cascade (20), whereas CSL-independent cellular responses, such as NICD-dependent activation of NF-κB (21), NICD-dependent inhibition of serine-protein kinase ATM (22) and NTMD-dependent activation of Ras-related C3 botulinum toxin substrate 1 (RAC1) (23), are defined as non-canonical Notch signaling cascades (Fig. 1).

NICDs undergo posttranslational modifications such as phosphorylation, ubiquitination and PARylation. Cyclin-dependent kinase (CDK)8-dependent phosphorylation of the NOTCH1 intracellular domain (NICD1) within the intracellular proline-, glutamate-, serine- and threonine-rich region leads to F-box/WD repeat-containing protein 7 (FBXW7)-mediated ubiquitination and proteasomal degradation (24,25), whereas ubiquitin carboxyl-terminal hydrolase 7-mediated deubiquitination stabilizes NOTCH1 receptors (26). SRC-dependent phosphorylation of NICD1 within the intracellular ankyrin repeat region represses Notch signaling through blockade of the NICD1-MAML interaction and degradation of NICD1 (27). AKT-dependent phosphorylation of NICD4 at S1495, S1847, S1865 and S1917 tethers NICD4 in the cytoplasm and represses NICD4-dependent transcription (28). MDM2-dependent NICD4 ubiquitination and E3 ubiquitin-protein ligase LNX (NUMB)-dependent NICD1 ubiquitination degrade NICDs and attenuate Notch signaling (29,30), whereas MDM2-dependent NICD1 ubiquitination does not degrade NICD1 and activates Notch signaling (31). Poly [ADP-ribose] polymerase tankyrase-1 (TNKS) PARylates NOTCH1, NOTCH2 and NOTCH3, and TNKS-dependent PARylation of NOTCH2 is required for nuclear translocation of the NICD (32). Posttranslational modifications of NICDs modulate their stability and intracellular localization to fine-tune intracellular Notch signaling.

Canonical Notch signals induce the upregulation of NICD/CSL-target genes (Fig. 1), such as BMI1 proto-oncogene polycomb ring finger (BMI1) (33,34), cyclin D1 (CCND1) (35,36), CD44 (37), CDKN1A (p21) (38,39), hes family bHLH transcription factor 1 (HES1) (40,41), hes family bHLH transcription factor 4 (HES4) (36,42), hes related family bHLH transcription factor with YRPW motif 1 (HEY1) (36,42,43), MYC (42,44,45), NOTCH3 (42,46), Notch regulated ankyrin repeat protein (NRARP) (36,41,42,47), nuclear factor erythroid 2 like 2 (48), olfactomedin 4 (OLFM4) (49), RE1 silencing transcription factor (REST) (41) and transcription factor 7 (TCF7) (50,51). Canonical Notch target genes are upregulated in a cellular context-dependent manner through dynamic patterns of Notch signaling activation, the epigenetic status of target genes and the availability of other transcription factors (16,52).

3. Notch signaling in tumor cells

Notch signaling molecules are frequently altered in T-ALL (80%) (53) and microsatellite-instable (MSI) or DNA polymerase-ε catalytic subunit A (POLE)-mutant subtypes of gastric and esophageal cancer (79%), colorectal cancer (70%) and uterine corpus endometrial cancer (64%) (54). Notch signaling is activated owing to gain-of-function (GoF) NOTCH alterations in T-ALL (55-57), chronic lymphocytic leukemia (58,59), diffuse large B cell lymphoma (60,61), mantle cell lymphoma (62), breast cancer (63-65) and non-small-cell lung cancer (NSCLC) (66) as well as loss-of-function (LoF) FBXW7 mutations in MSI or POLE-mutant cancers and hematological malignancies (53,54) (Fig. 2). By contrast, Notch signaling is inactivated as a result of LoF NOTCH alterations in cutaneous squamous cell carcinoma (67), head and neck squamous cell carcinoma (HNSCC) (68,69), esophageal squamous cell carcinoma (70,71) and SCLC (72) (Fig. 2).

Transcriptional or epigenetic alterations also dysregulate Notch signaling in the absence of genetic alterations in the Notch signaling components (Fig. 2). Oncogenic Notch signaling is reinforced due to NOTCH3 upregulation through ETS-related transcription factor ELF3-dependent transcription in KRAS-mutant lung adenocarcinoma (73); JAG1 upregulation through CpG hypomethylation in renal cell carcinoma (74); and upregulation of JAG1, MAML2, NOTCH1, NOTCH2 and NOTCH3, partially through increased histone H3K27 acetylation, in neuroblastoma (75). Tumor-suppressive Notch signaling is inactivated in Ewing's sarcoma due to repression of JAG1 by RNA binding protein EWS-friend leukemia integration 1 transcription factor fusion protein (76) and repression of NOTCH1 and REST through decreased H3K27 acetylation in SCLC (77).

Notch signaling activation promotes tumor cell proliferation or survival and in vivo tumorigenesis through: i) Direct upregulation of CCND1 (35) and MYC (44); ii) HES1-mediated CDKN1B (p27) repression and subsequent cellular proliferation (78); iii) HES1-mediated dual specificity phosphatase 1 repression and subsequent ERK activation (79); iv) HES1-mediated phosphatase and tensin homolog repression and subsequent AKT signaling activation (80); and v) HES1-mediated STAT3 activation (81,82) and CSL-independent, NF-κB-dependent interleukin 6 (IL6) upregulation, and subsequent JAK-STAT signaling activation (83). By contrast, Notch signaling activation blocks tumor cell proliferation or survival and in vivo tumorigenesis through: i) Direct upregulation of CDKN1A (38,39); ii) HES1-mediated GLI family zinc finger 1 repression (84); iii) HEY1-mediated snail family transcriptional repressor 2 and twist family bHLH transcription factor 1 repression, and subsequent mesenchymal-to-epithelial transition (85); and iv) HEY1-mediated IL6 downregulation and subsequent depletion of cancer stem cells (86). Because Notch signals drive lateral induction as well as lateral inhibition to fine-tune organ development and homeostasis (17,87,88), bifunctional cellular responses are a common feature of Notch signaling during embryogenesis, adult tissue homeostasis and tumorigenesis.

Oncogenic Notch signaling is activated in NSCLC owing to GoF NOTCH1 mutations or ELF3-dependent NOTCH3 upregulation (66,73), whereas tumor-suppressive Notch signaling is inactivated in SCLC owing to LoF NOTCH1 mutations or epigenetic NOTCH1 repression (72,77). In HNSCC, tumor-suppressive Notch signaling is inactivated owing to LoF NOTCH1 mutations, but oncogenic Notch signaling is activated by JAG1, JAG2 or NOTCH3 upregulation (69,89). Tumor-suppressive Notch signaling is advantageous for maintaining a non-cancerous esophagus in middle-aged or elderly individuals (71), whereas oncogenic Notch signaling promotes the later stages of T-cell leukemogenesis (57) and B-cell lymphomagenesis (61). Because Notch signals intrinsically exert both oncogenic and tumor-suppressive effects (Fig. 1), epigenetic silencing or genetic inactivation of anti-tumorigenic Notch target genes may transfer the growth advantage from LoF Notch mutants to GoF Notch mutants.

4. Notch signaling in the tumor microenvironment

The tumor microenvironment comprises a heterogeneous population of cancer cells, cancer-associated fibroblasts (CAFs), endothelial cells, mesenchymal stem/stromal cells (MSCs), pericytes, peripheral neurons and immune cells (90-92) (Fig. 3). Single-cell RNA sequencing (scRNAseq) revealed seven subgroups of fibroblasts, six subgroups of endothelial cells and 30 subgroups of immune cells in NSCLC (93), and four subtypes of cancer-associated fibroblasts in mouse mammary tumors (94). Cancerous and non-cancerous cells communicate via growth factors, cytokines and extracellular vesicles for paracrine signaling, and via membrane-type ligand/receptor pairs for juxtacrine signaling (3,95-97). These intercellular communications turn the anti-tumor microenvironment into a pro-tumor microenvironment through 'omics reprogramming' (98), which includes epigenetic changes (99), epithelial-to-mesenchymal transition (100), immunoediting (101) and vascular remodeling (102).

Notch4 (Int3), fibroblast growth factor (Fgf) 3 (Int2), Fgf4, R-spondin (Rspo) 2 (Int7), Rspo3, Wnt1 (Int1) and Wnt3 (Int4) are proto-oncogenes that are activated by mouse mammary tumor virus (MMTV) (103-108). Notch signaling is required for the CSL-dependent expression of FGF7, FGF9, FGF10, FGF18, WNT1, WNT2 and WNT3 in dermal fibroblasts (39), while RSPO2 and RSPO3 interact with LGR4/5/6 to potentiate WNT signaling through Frizzled receptors (109,110). WNT signals enhance Notch signaling through JAG1 and NOTCH2 upregulation (111,112) but repress Notch signaling through NUMB and prospero homeobox 1 upregulation (113,114). Notch signals enhance β-catenin/LEF1 signaling via NRARP upregulation (47,115), but repress WNT/β-catenin signaling through OLFM4 upregulation (49,116). Notch and WNT signals converge on BMI1 and TCF7 to maintain slow-cycling cancer stem cells, partially through BMI1-induced telomerase reverse transcriptase upregulation and TCF7-induced CDKN2 upregulation, and on CCND1 and MYC to promote tumor proliferation (34,50,51,117-121). Colorectal cancer stem cells diverge into Notch- and WNT-dependent populations, and Notch signals may not be essential for bulk tumorigenesis (122,123). Notch signaling cascades crosstalk with FGF and WNT signaling cascades to orchestrate the tumor microenvironment for the maintenance of cancer stem cells.

Tumor angiogenesis is characterized by excessive endothelial sprouting from preexisting blood vessels, which leads to overgrowth of randomly organized and leaky tumor vessels (124-126). Vascular endothelial growth factor (VEGFA) signaling through VEGF receptor 2 (VEGFR2) (KDR) and neuropilin-1 (NRP1) receptors on endothelial tip cells drives vascular sprouting and DLL4 upregulation, and DLL4 signaling through Notch receptors on endothelial stalk cells restricts angiogenic sprouting and proliferation through downregulation of VEGFR2 and NRP1 (127,128). By contrast, Notch signaling induces JAG1 upregulation to antagonize the DLL4-dependent 'stalk' phenotype, and promote endothelial sprouting and proliferation (129,130). NICD1-dependent Notch signaling activation in endothelial cells promotes lung metastasis (131), but that in hepatic endothelial cells represses liver metastasis (132). Thus, Notch signaling regulates tumor angiogenesis and metastasis in a context-dependent manner.

Notch signals are involved in the development and homeostasis of immune cells: JAG1-Notch, DLL4-Notch1 and DLL1-Notch2 signals promote the self-renewal of long-term hematopoietic stem cells, differentiation of early T-lymphocyte progenitors and differentiation of marginal zone B lymphocytes, respectively (133,134); DLL1/4 and JAG1/2 signals induce the differentiation of naïve T lymphocytes into Th1 and Th2 cells, respectively (135,136); DLL1 and JAG1 signals promote the differentiation of tumor-associated macrophages (TAMs) into M1- and M2-like phenotypes, respectively (137,138); DLL1 or JAG1 on MSCs and JAG2 on hematopoietic progenitor cells induce the expansion of regulatory T (Treg) cells (139-141); and DLL4 on dendritic cells promotes Treg differentiation (142). By contrast, Notch-related immunological reprogramming in the tumor microenvironment may be more complex; scRNAseq revealed 20 subsets of T lymphocytes, including circulating Treg cells, non-cancerous tissue-infiltrating Treg cells and cancerous tissue-infiltrating Treg cells (143). For example, Notch-mediated immune regulation in the hypoxic tumor microenvironment is potentiated by the interaction between NICD and hypoxia-induced hypoxia inducible factor-1α, and is modulated by the crosstalk with the FGF, Hedgehog, transforming growth factor (TGF)-β, VEGF and WNT signaling cascades (102,124,125,144-147). Notch1 signaling elicited immune evasion through TGF-β upregulation and accumulation of myeloid-derived suppressor cells (MDSCs) and Treg cells in a mouse xenograft model with B16 melanoma cells (148), and through upregulation of cytotoxic T-lymphocyte protein 4, lymphocyte activation gene 3 protein, programmed cell death protein 1 and hepatitis A virus cellular receptor 2, and accumulation of MDSCs, TAMs and Treg cells, in an engineered mouse model of HNSCC (149).

5. Therapeutics targeting Notch signaling cascades

Investigational drugs that target Notch signaling cascades are classified as follows: i) Small-molecule γ-secretase inhibitors that block the final step of ligand-induced processing of Notch receptors; ii) biologics, including mAbs, ADCs, bsAbs and CAR-Ts, that bind to the extracellular region of Notch ligands or receptors; iii) ADAM17 inhibitors that block the initial step of ligand-induced processing of Notch receptors; and iv) NICD protein-protein-interaction inhibitors that block the NICD-dependent transcription of Notch target genes (Table I).

Table I

Notch-targeted therapeutics.

Table I

Notch-targeted therapeutics.

ClassDrugAliasMechanism of actionStage of drug development(Refs.)
GSIAL101BMS-906024Inhibition of S3 cleavagePhase II (registration no. NCT03691207; GoF-Notch ACC; Recruiting)(150)
CrenigacestatLY3039478Inhibition of S3 cleavagePhase I (registration no. NCT01695005; advanced cancer; completed)(151)
MRK-560Inhibition of S3 cleavagePreclinical study (PSEN1-sublass GSI inhibitor for T-ALL)(152)
NirogacestatPF-03084014Inhibition of S3 cleavagePhase III (registration no. NCT03785964; desmoid tumors; recruiting)(153,154)
RO4929097Inhibition of S3 cleavagePhase II (Multiple trials failed, insufficient or terminated)(155,156)
mAbDemcizumabOMP-21M18Anti-DLL4 mAbPhase II (registration no. NCT02259582; w/Chemo; NSCLC; completed)(160)
EnoticumabREGN421Anti-DLL4 mAbPhase I (registration no. NCT00871559; solid tumors; completed)(161)
MEDI0639Anti-DLL4 mAbPhase I (registration no. NCT01577745; solid tumors; completed)(162)
BrontictuzumabOMP-52M51Anti-NOTCH1 mAbPhase I (registration no. NCT01778439; solid tumors; completed)(163)
TarextumabOMP-59R5Anti-NOTCH2/3 mAbPhase II (registration no. NCT01647828; w/Chemo; Panc; completed)(164,165)
15D11Anti-JAG1 mAbPreclinical study(220)
ADCRovalpituzumab tesirineRova-T, SC16LD6.5Anti-DLL3 ADCPhase III (registration no. NCT03033511; SCLC; recruiting); Phase III (registration no. NCT03061812; DLL3-high SCLC; active NR)(170-172)
PF-06650808Anti-NOTCH3 ADCPhase I (registration no. NCT02129205; solid tumors; terminated)(173)
bsAbAMG 757Anti-DLL3/CD3 bsAbPhase I (registration no. NCT03319940; SCLC; recruiting)(174)
ABT-165Anti-DLL4/VEGF bsAbPhase II (registration no. NCT03368859; w/Chemo; CRC; recruiting)(175)
NavicixizumabOMP-305B83Anti-DLL4/VEGF bsAbPhase I (registration no. NCT02298387; solid tumors; completed)(176)
CT16Anti-NOTCH2/3/EGFR bsAbPreclinical study(177)
PTG12Anti-NOTCH2/3/EGFR bsAbPreclinical study(178)
CAR-TsAMG 119DLL3-binding CAR-TsPhase I (registration no. NCT03392064; SCLC; active NR)(179)
OthersZLDI-8ADAM17 inhibitorPreclinical study(221)
CB-103NICD PPI inhibitorPhase I/II (registration no. NCT03422679; cancer; recruiting)(222)
SAHM1NICD PPI inhibitorPreclinical study(223)

[i] ACC, adenoid cystic carcinoma; Active NR, active, not recruiting; ADC, antibody-drug conjugate; bsAb, bispecific antibody or biologic; CAR-Ts, chimeric antigen receptor-modified T cells; CRC, colorectal cancer; GSI, γ-secretase inhibitor; mAb, monoclonal antibody; NICD, Notch intracellular domain; NSCLC, non-small-cell lung cancer; Panc, pancreatic cancer; PPI, protein-protein interaction; PSEN1, presenilin-1; SCLC, small-cell lung cancer; T-ALL, T-cell acute lymphoblastic leukemia; w/, with; GoF, gain-of-function; DLL, delta-like canonical Notch ligand; JAG, jagged canonical Notch ligand; VEGF, vascular endothelial growth factor; EGFR, epidermal growth factor receptor; ADAM17, disintegrin and metalloproteinase domain-containing protein.

γ-Secretase inhibitors, such as AL101 (150), crenigacestat (151), MRK-560 (152), nirogacestat (153,154) and RO4929097 (155,156), are investigational Notch pathway inhibitors. AL101, crenigacestat, nirogacestat and RO4929097 were tolerated in phase I clinical trials with common adverse effects, such as diarrhea, fatigue, nausea and vomiting (150,151,153,155), whereas MRK-560, which selectively targets presenilin-1-containing γ-secretase complexes, is a next-generation γ-secretase inhibitor with decreased gastrointestinal toxicities (152). Multiple phase II clinical trials of RO4929097 (registration nos. NCT01116687, NCT01120275, NCT01175343 and NCT01232829) failed, had insufficient results or were terminated because of limited anti-tumor activity, partially driven by cytochrome P450 3A4-mediated drug metabolism (155,156). Combination therapy is a rational strategy to enhance the clinical benefits of γ-secretase inhibitors, because bypassing the activation of receptor tyrosine kinases (RTKs) (157,158) and the RAS-MEK-ERK (159), PI3K-AKT (80) and Hedgehog-GLI (84) signaling cascades elicits resistance to γ-secretase inhibitors. Prescription to strong responders is another rational strategy to enhance the clinical benefits of γ-secretase inhibitors. A phase III clinical trial of nirogacestat for desmoid tumor patients (registration no. NCT03785964) is in progress based on objective response rates (ORRs) of ~70 and ~30% in phase I (registration no. NCT00878189) and phase II (registration no. NCT01981551) clinical trials, respectively (153,154).

Antibody drugs that can selectively block Notch ligands or receptors have been predicted to be an optimal choice for cancer therapy compared with γ-secretase inhibitors for pan-Notch signaling blockade. Anti-DLL4 mAbs (demcizumab, enoticumab and MEDI0639) (160-162), an anti-NOTCH1 mAb (brontictuzumab) (163) and an anti-NOTCH2/3 mAb (tarextumab) (164,165) have been investigated in phase I clinical trials for the treatment of patients with cancer (Table I), and were relatively well tolerated with common adverse effects, including diarrhea, fatigue and nausea. However, because DLL4-NOTCH signaling in endothelial cells (127,128) and DLL4-NOTCH3 signaling in pericytes (5) mediate cardiovascular homeostasis, anti-DLL4 and anti-NOTCH2/3 mAbs elicit cardiovascular toxicities, such as hypertension, acute myocardial infarction, left ventricular dysfunction and peripheral edema. The ORRs of monotherapy with anti-DLL4, anti-NOTCH1 and anti-NOTCH2/3 mAbs were <5% (160-165).

ADC, bsAb and CAR-T technologies (166-169) have been applied to enhance the benefits of therapeutic mAbs in patients with cancer. Notch-related investigational biologics include ADCs targeting DLL3 [rovalpituzumab tesirine (Rova-T)] (170-172) and NOTCH3 (PF-06650808) (173); bsAbs targeting DLL3/CD3 (AMG 757) (174), DLL4/VEGF (ABT-165 and navicixizumab) (175,176) and NOTCH2/3/EGFR (CT16 and PTG12) (177,178); and CAR-Ts targeting DLL3 (AMG 119) (179) (Table I). A phase I clinical trial of the anti-DLL4/VEGF bsAb navicixizumab in 66 patients with solid tumors (registration no. NCT02298387) showed four partial responses (PRs) in the entire cohort and three PRs among 11 patients with ovarian cancer, accompanied by adverse events such as systemic hypertension (58%) and pulmonary hypertension (18%) (176); in addition, a phase I clinical trial of the anti-NOTCH3 ADC PF-06650808 in patients with breast cancer and other solid tumors (registration no. NCT02129205) revealed a manageable safety profile and three PRs among 40 participants (173). By contrast, a phase I clinical trial of the anti-DLL3 ADC Rova-T in 74 patients with SCLC and eight patients with large-cell neuroendocrine tumors (registration no. NCT01901653) demonstrated ORRs of 17% (11/65) in the entire cohort and 38% (10/26) among DLL3-high patients, with adverse events such as thrombocytopenia and pleural effusion (171). Preliminary analysis of a phase II clinical trial of Rova-T in patients with SCLC (registration no. NCT02674568) showed an ORR of 21.6% (58/266), with manageable toxicities (172). Currently, phase III clinical trials of Rova-T for the treatment of SCLC patients (registration nos. NCT03033511 and NCT03061812) are ongoing. Regarding DLL3, phase I clinical trials of the anti-DLL3/CD3 bsAb AMG 757 (registration no. NCT03319940) and DLL3-targeting CAR-Ts AMG 119 (registration no. NCT03392064) are also in progress. Compared with DLL4 and NOTCH3, DLL3 is an ideal target for ADCs, bsAbs and CAR-Ts, because DLL3 is upregulated in SCLC and other neuroendocrine tumors, repressing Notch signaling and reciprocally upregulating REST to maintain the neuroendocrine phenotype (41,170,180).

6. Perspectives on Notch-targeted precision oncology

ADCs or CAR-Ts targeting RTKs (Table II) and other trans-membrane or GPI-anchored proteins (Table III) are popular topics in clinical oncology. Anti-CD19 CAR-Ts (axicabtagene ciloleucel and tisagenlecleucel) (181,182), an anti-CD22 ADC (inotuzumab ozogamicin) (183), an anti-CD30 ADC (brentuximab vedotin) (184) and an anti-CD79B ADC (polatuzumab vedotin) (185) have been approved by the US Food and Drug Administration for the treatment of patients with hematological malignancies, and an anti-HER2 ADC (trastuzumab emtansine) (186) has been approved for the treatment of patients with breast cancer (Fig. 4). Trastuzumab-based ADCs with distinct linkers and payloads (trastuzumab deruxtecan and trastuzumab duocarmazine) (187,188); other ADCs targeting epidermal growth factor receptor (EGFR) (189), folate receptor-α (190), NECTIN4 (191) and tumor-associated calcium signal transducer 2 (192); and CAR-Ts targeting tumor necrosis factor receptor superfamily member 17 (193) are also in phase III clinical trials. An anti-CD205 ADC that targets mesenchymal tumor cells and CAFs (194) and anti-Claudin-18.2 CAR-Ts that showed an ORR of 36% (4/11) in patients with gastric or pancreatic cancer (195) are cutting-edge biologics in early-stage clinical trials. ADCs and CAR-Ts (Tables II and III) could alter the therapeutic scheme for refractory solid tumors, especially peritoneal dissemination from diffuse-type gastric cancer, ovarian cancer and pancreatic cancer.

Table II

ADCs and CAR-Ts targeting RTKs.

Table II

ADCs and CAR-Ts targeting RTKs.

TargetTypeDrug nameAliasStage of drug development(Refs.)
ALKADCCDX-0125-TEIPreclinical study (Rodent)(224)
AXLADCEnapotamab vedotinAXL-107-MMAEPhase I/II (registration no. NCT02988817; solid tumors; Recruiting)(225)
DDR1ADCT4H11-DM4Preclinical study (Rodent)(226)
EGFRADCDepatuxizumab mafodotinABT-414Phase II/III (registration no. NCT02573324; EGFR+ Glio; active NR)(189)
FGFR2ADCBAY 1179470Phase I (registration no. NCT02368951; solid tumors; completed in 2016)(227)
FGFR3ADCLY3076226Phase I (registration no. NCT02529553; cancer; completed in 2018)(228)
FLT3ADCASP1235AGS62P1Phase I (registration no. NCT02864290; AML; recruiting)(229)
HER2ADCTrastuzumab emtansineT-DM1FDA approval (HER2+ Breast)(186)
HER2ADCTrastuzumab deruxtecanDS-8201aPhase III (registration no. NCT03529110; HER2+ Breast; recruiting)(187)
HER2ADCTrastuzumab duocarmazineSYD985Phase III (registration no. NCT03262935; HER2+ Breast; recruiting)(188)
HER2ADCMI130004Preclinical study (rodent; long-lasting anti-tumor effects)(230)
HER3ADCU3-1402Phase I/II (registration no. NCT02980341; HER3+ Breast; recruiting)(231)
KITADCLOP628Phase I (registration no. NCT02221505; KIT+ Cancers; terminated in 2015)(232)
METADCTelisotuzumab vedotinTeliso-V or ABBV-399Phase II (registration no. NCT03539536; MET+ NSCLC; recruiting)(233)
PTK7ADCCofetuzumab pelidotinPF-06647020Phase I (registration no. NCT02222922; solid tumors; active NR)(234)
RETADCY078-DM1Preclinical study (rodent & primate; on-target neuropathy)(235)
RONADCH-Zt/g4-MMAEPreclinical study (rodent & primate)(236)
ROR1CAR-TsROR1 CAR-TsPreclinical study (rodent & primate)(237)

[i] Active NR, active, not recruiting; ADC, antibody-drug conjugate; AML, acute myeloid leukemia; Breast, breast cancer; CAR-Ts, chimeric antigen receptor-modified T cells; EGFR+, epidermal growth factor receptor-amplified; Glio, glioblastoma or gliosarcoma; NSCLC, non-small-cell lung cancer; FDA, Food and Drug Administration; RTK, receptor tyrosine kinase; ALK, ALK tyrosine kinase receptor; AXL, tyrosine-protein kinase receptor UFO; DDR1, epithelial discoidin domain-containing receptor 1; FGFR, fibroblast growth factor receptor; FLT3, receptor-type tyrosine-protein kinase FLT3; KIT, mast/stem cell growth factor receptor Kit; PTK7, inactive tyrosine-protein kinase 7; RET, proto-oncogene tyrosine-protein kinase receptor Ret; RON, macrophage-stimulating protein receptor; ROR1, inactive tyrosine-protein kinase transmembrane receptor ROR1.

Table III

ADCs and CAR-Ts targeting transmembrane or GPI-anchored proteins other than DLL3, NOTCH3 and RTKs.

Table III

ADCs and CAR-Ts targeting transmembrane or GPI-anchored proteins other than DLL3, NOTCH3 and RTKs.

TargetTypeDrug nameAliasStage of drug development(Refs.)
BCMAADCGSK2857916Phase II (registration no. NCT03525678; multiple myeloma; active NR)(238)
BCMACAR-TsBb2121Phase III (registration no. NCT03651128; multiple myeloma; recruiting)(193)
CD19CAR-TsAxicabtagene ciloleucelKTE-C19FDA approval (B-cell NHL)(181)
CD19CAR-Ts TisagenlecleucelCTL019FDA approval (B-cell ALL & NHL)(182)
CD22ADCInotuzumab ozogamicinCMC-544FDA approval (B-cell ALL)(183)
CD30ADCBrentuximab vedotinSGN-35FDA approval (ALCL, HL, mycosis fungoides & PTCL)(184)
CD33ADCGemtuzumab ozogamicinCMA-676FDA approval (AML) and subsequent withdrawal(239)
CD56ADCLorvotuzumab mertansineIMGN901Phase II (registration no. NCT02452554; pediatric tumors; active NR)(240)
CD79BADCPolatuzumab vedotinDCDS4501AFDA approval (diffuse large B-cell lymphoma)(185)
CD142ADCTisotumab vedotinTF-ADCPhase I/II (registration no. NCT02001623; solid tumors; completed in 2018)(241)
CD205ADCMEN1309OBT076Phase I (registration no. NCT03403725; solid tumors; recruiting)(194)
CEACAM5ADCLabetuzumab govitecanIMMU-130Phase I/II (registration no. NCT01605318; colorectal cancer; completed in 2017)(242)
CLDN18ADCAnti-CLDN18.2 ADCPreclinical study (rodent)(243)
CLDN18CAR-TsCAR-CLDN18.2Phase I (registration no. NCT03159819; Gas & Panc; recruiting)(195)
FOLR1ADCMirvetuximab soravtansineIMGN853Phase III (registration no. NCT02631876; ovary; active NR)(190)
GFRA1ADC hu-6D3.v5-vcMMAEPreclinical study (rodent & primate)(244)
GPNMBADCGlembatumumab vedotinCDX-011Phase II (registration no. NCT02302339; melanoma; terminated in 2018)(245)
LGR5ADC Anti-LGR5-mc-vc-PAB-MMAEPreclinical study (rodent)(246)
LRRC15ADCSamrotamab vedotinABBV-085Phase I (registration no. NCT02565758; solid tumors; completed in 2019)(247)
LYPD3ADCLupartumab amadotinBAY 1129980Phase I (registration no. NCT02134197; solid tumors; completed in 2018)(248)
MSLNADCAnetumab ravtansineBAY 94-9343Phase II (registration no. NCT03023722; Panc; active NR)(249)
NECTIN4ADCEnfortumab vedotinASG-22MEPhase III (registration no. NCT03474107; urothelial cancer; recruiting)(191)
SLC34A2ADCLifastuzumab vedotinDNIB0600APhase II (registration no. NCT01991210; ovarian cancer; completed in 2016)(250)
SLC39A6ADCLadiratuzumab vedotinSGN-LIV1APhase I/II (registration no. NCT03310957; TNBC; recruiting)(251)
TROP2ADCSacituzumab govitecanIMMU-132Phase III (registration no. NCT02574455; TNBC; recruiting)(192)

[i] Active NR, active, not recruiting; ADC, antibody-drug conjugate; ALCL, anaplastic large cell lymphoma; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; Breast, breast cancer; CAR-Ts, chimeric antigen receptor-modified T cells; CD142, Tissue factor; Gas, gastric cancer; HL, Hodgkin lymphoma; MSLN, Mesothelin; NHL, non-Hodgkin lymphoma; Ovary, ovarian, fallopian tube or primary peritoneal cancer; Panc, pancreatic cancer; PTCL, peripheral T-cell lymphoma; RTK, receptor tyrosine kinase, SCLC, small-cell lung cancer; TNBC, triple-negative breast cancer; FDA, Food and Drug Administration; BCMA, tumor necrosis factor receptor superfamily member 17; CEACAM5, carcinoembryonic antigen-related cell adhesion molecule 5; CLDN18, Claudin 18.2; FOLR1, folate receptor-α; GFRA1, GDNF family receptor α1; GPNMB, transmembrane glycoprotein NMB; LGR5, leucine-rich repeat-containing G-protein coupled receptor 5; LRRC15, leucine-rich repeat-containing protein 15; LYPD3, Ly6/PLAUR domain-containing protein 3; MSLN, mesothelin; SLC34A2, sodium-dependent phosphate transport protein 2B; SLC39A6, zinc transporter SIP6; TROP2, tumor-associated calcium signal transducer 2.

Repression of targeted antigens owing to the intratu-moral heterogeneity and omics reprogramming of tumor cells is a common mechanism of resistance to ADCs and CAR-Ts (98,196,197). Clinical trials of ADCs in patients with solid tumors have produced disappointing results, owing to a narrow therapeutic window and unavoidable therapeutic resistance or recurrence (Tables II and III). Recruitment of new patients for the randomized phase III clinical trial of Rova-T in patients with SCLC (registration no. NCT03061812) was halted owing to shorter overall survival times in the Rova-T treatment group than in the topotecan treatment group (12). LoF NOTCH1 mutations that decrease DLL3 dependence to suppress Notch signaling might lead to intrinsic resistance to Rova-T, whereas trans-differentiation from DLL3-high SCLC to DLL3-low SCLC or NSCLC might elicit acquired resistance to Rova-T. To enhance the clinical benefits of Rova-T in patients with SCLC, the mechanisms of resistance and biomarkers of responders should be elucidated by monitoring DLL3 expression, NOTCH mutations and tumor phenotypes before, during and after Rova-T therapy.

Clinical genomic tests using panel-based next-generation sequencing are utilized to match approved marketed drugs or investigational drugs to cancer patients in clinical trials in the era of precision oncology (198-200) (Fig. 5). These up-to-date genomic tests, which detect alterations in 400-500 cancer-related genes, but not out-of-date genomic tests, which detect many fewer cancer-related genes, can be reliably applied to diagnose tumor mutational burden-high cancers that predict responders to immune checkpoint inhibitors and non-responders to EGFR inhibitors (201-204). By contrast, because of their optimization for the major genetic alterations in various human cancer types, panel-based genomic tests cannot detect rare genetic alterations, promoter/enhancer mutations and epigenetic alterations that elicit aberrant activation of Notch and other oncogenic signaling pathways. Genomic tests that detect GoF mutations in the NOTCH1, NOTCH2, NOTCH3 and NOTCH4 genes, as well as mRNA in situ hybridization and immunohistochemical analyses that detect overexpression of Notch family receptors, would enhance the benefits of Notch pathway inhibitors, such as blocking mAbs and γ-secretase inhibitors, through successful positive selection of putative responders.

Whole-genome sequencing, as well as wholeexome sequencing plus transcriptome analysis, is applied for the exploration of unknown cancer drivers, and the development of novel therapeutics for known but intractable targets with the aid of human intelligence, cognitive computing and artificial intelligence in basic and translational oncology (205-208). Moreover, artificial intelligence is also applied for computer-aided diagnostic approaches (209,210), such as chest computed tomography (211), dermoscopy (212), gastrointestinal endoscopy (213), mammography (214) and histopathological diagnosis (215-218). To avoid the lack of transparency associated with black box artificial intelligence based on deep learning technologies, the development of explainable artificial intelligence is necessary (219). Construction of a Notch-related knowledge base via human intelligence, explainable artificial intelligence, and cognitive computing based on natural language processing and text mining (Fig. 6) would promote the clinical application of Notch-targeted therapeutics in the era of omics-based precision medicine.

Funding

This study was supported in part by a grant-in-aid from Masaru Katoh's Fund for the Knowledge-Base Project.

Availability of data and materials

Not applicable.

Authors' contributions

MasukoK and MasaruK contributed to the conception of the study, performed the literature search and wrote the manuscript. MasukoK prepared the tables. MasaruK prepared the figures. All the 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.

Acknowledgments

Not applicable.

References

1 

Guruharsha KG, Kankel MW and Artavanis-Tsakonas S: The Notch signalling system: Recent insights into the complexity of a conserved pathway. Nat Rev Genet. 13:654–666. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Bray SJ: Notch signalling in context. Nat Rev Mol Cell Biol. 17:722–735. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Meurette O and Mehlen P: Notch signaling in the tumor micro-environment. Cancer Cell. 34:536–548. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Siebel C and Lendahl U: Notch signaling in development, tissue homeostasis, and disease. Physiol Rev. 97:1235–1294. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Wimmer RA, Leopoldi A, Aichinger M, Wick N, Hantusch B, Novatchkova M, Taubenschmid J, Hämmerle M, Esk C, Bagley JA, et al: Human blood vessel organoids as a model of diabetic vasculopathy. Nature. 565:505–510. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Ranganathan P, Weaver KL and Capobianco AJ: Notch signalling in solid tumours: A little bit of everything but not all the time. Nat Rev Cancer. 11:338–351. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Ntziachristos P, Lim JS, Sage J and Aifantis I: From fly wings to targeted cancer therapies: A centennial for notch signaling. Cancer Cell. 25:318–334. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Aster JC, Pear WS and Blacklow SC: The varied roles of Notch in cancer. Annu Rev Pathol. 12:245–275. 2017. View Article : Google Scholar

9 

Nowell CS and Radtke F: Notch as a tumour suppressor. Nat Rev Cancer. 17:145–159. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Espinoza I and Miele L: Notch inhibitors for cancer treatment. Pharmacol Ther. 139:95–110. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, Yang SX and Ivy SP: Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: Clinical update. Nat Rev Clin Oncol. 12:445–464. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Owen DH, Giffin MJ, Bailis JM, Smit MD, Carbone DP and He K: DLL3: An emerging target in small cell lung cancer. J Hematol Oncol. 12:612019. View Article : Google Scholar : PubMed/NCBI

13 

D'Souza B, Miyamoto A and Weinmaster G: The many facets of Notch ligands. Oncogene. 27:5148–5167. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Kangsamaksin T, Murtomaki A, Kofler NM, Cuervo H, Chaudhri RA, Tattersall IW, Rosenstiel PE, Shawber C and Kitajewski J: NOTCH decoys that selectively block DLL/NOTCH or JAG/NOTCH disrupt angiogenesis by unique mechanisms to inhibit tumor growth. Cancer Discov. 5:182–197. 2015. View Article : Google Scholar

15 

Kakuda S and Haltiwanger RS: Deciphering the Fringe-mediated Notch code: Identification of activating and inhibiting sites allowing discrimination between ligands. Dev Cell. 40:193–201. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Nandagopal N, Santat LA, LeBon L, Sprinzak D, Bronner ME and Elowitz MB: Dynamic ligand discrimination in the Notch signaling pathway. Cell. 172:869–880.e19. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Sjöqvist M and Andersson ER: Do as I say, Not(ch) as I do: Lateral control of cell fate. Dev Biol. 447:58–70. 2019. View Article : Google Scholar

18 

Lambrecht BN, Vanderkerken M and Hammad H: The emerging role of ADAM metalloproteinases in immunity. Nat Rev Immunol. 18:745–758. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Yang G, Zhou R, Zhou Q, Guo X, Yan C, Ke M, Lei J and Shi Y: Structural basis of Notch recognition by human γ-secretase. Nature. 565:192–197. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Kopan R and Ilagan MX: The canonical Notch signaling pathway: Unfolding the activation mechanism. Cell. 137:216–233. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Vilimas T, Mascarenhas J, Palomero T, Mandal M, Buonamici S, Meng F, Thompson B, Spaulding C, Macaroun S, Alegre ML, et al: Targeting the NF-kappaB signaling pathway in Notch1-induced T-cell leukemia. Nat Med. 13:70–77. 2007. View Article : Google Scholar

22 

Vermezovic J, Adamowicz M, Santarpia L, Rustighi A, Forcato M, Lucano C, Massimiliano L, Costanzo V, Bicciato S, Del Sal G and d'Adda di Fagagna F: Notch is a direct negative regulator of the DNA-damage response. Nat Struct Mol Biol. 22:417–424. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Polacheck WJ, Kutys ML, Yang J, Eyckmans J, Wu Y, Vasavada H, Hirschi KK and Chen CS: A non-canonical Notch complex regulates adherens junctions and vascular barrier function. Nature. 552:258–262. 2017. View Article : Google Scholar : PubMed/NCBI

24 

O'Neil J, Grim J, Strack P, Rao S, Tibbitts D, Winter C, Hardwick J, Welcker M, Meijerink JP, Pieters R, et al: FBW7 mutations in leukemic cells mediate NOTCH pathway activation and resistance to γ-secretase inhibitors. J Exp Med. 204:1813–1824. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Wang Z, Liu P, Inuzuka H and Wei W: Roles of F-box proteins in cancer. Nat Rev Cancer. 14:233–247. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Shan H, Li X, Xiao X, Dai Y, Huang J, Song J, Liu M, Yang L, Lei H, Tong Y, et al: USP7 deubiquitinates and stabilizes NOTCH1 in T-cell acute lymphoblastic leukemia. Signal Transduct Target Ther. 3:292018. View Article : Google Scholar : PubMed/NCBI

27 

LaFoya B, Munroe JA, Pu X and Albig AR: Src kinase phosphorylates Notch1 to inhibit MAML binding. Sci Rep. 8:155152018. View Article : Google Scholar : PubMed/NCBI

28 

Ramakrishnan G, Davaakhuu G, Chung WC, Zhu H, Rana A, Filipovic A, Green AR, Atfi A, Pannuti A, Miele L and Tzivion G: AKT and 143-3 regulate Notch4 nuclear localization. Sci Rep. 5:87822015. View Article : Google Scholar

29 

Sun Y, Klauzinska M, Lake RJ, Lee JM, Santopietro S, Raafat A, Salomon D, Callahan R and Artavanis-Tsakonas S: Trp53 regulates Notch 4 signaling through Mdm2. J Cell Sci. 124:1067–1076. 2011. View Article : Google Scholar : PubMed/NCBI

30 

McGill MA and McGlade CJ: Mammalian Numb proteins promote Notch1 receptor ubiquitination and degradation of the Notch1 intracellular domain. J Biol Chem. 278:23196–23203. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Pettersson S, Sczaniecka M, McLaren L, Russell F, Gladstone K, Hupp T and Wallace M: Non-degradative ubiquitination of the Notch1 receptor by the E3 ligase MDM2 activates the Notch signalling pathway. Biochem J. 450:523–536. 2013. View Article : Google Scholar

32 

Bhardwaj A, Yang Y, Ueberheide B and Smith S: Whole proteome analysis of human tankyrase knockout cells reveals targets of tankyrase-mediated degradation. Nat Commun. 8:22142017. View Article : Google Scholar : PubMed/NCBI

33 

Schaller MA, Logue H, Mukherjee S, Lindell DM, Coelho AL, Lincoln P, Carson WF IV, Ito T, Cavassani KA, Chensue SW, et al: Delta-like 4 differentially regulates murine CD4 T cell expansion via BMI1. PLoS One. 5:e121722010. View Article : Google Scholar : PubMed/NCBI

34 

López-Arribillaga E, Rodilla V, Pellegrinet L, Guiu J, Iglesias M, Roman AC, Gutarra S, González S, Muñoz-Cánoves P, Fernández- Salguero P, et al: Bmi1 regulates murine intestinal stem cell proliferation and self-renewal downstream of Notch. Development. 142:41–50. 2015. View Article : Google Scholar

35 

Ronchini C and Capobianco AJ: Induction of cyclin D1 transcription and CDK2 activity by Notch(ic): Implication for cell cycle disruption in transformation by Notch(ic). Mol Cell Biol. 21:5925–5934. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Tanis KQ, Podtelezhnikov AA, Blackman SC, Hing J, Railkar RA, Lunceford J, Klappenbach JA, Wei B, Harman A, Camargo LM, et al: An accessible pharmacodynamic transcriptional biomarker for Notch target engagement. Clin Pharmacol Ther. 99:370–380. 2016. View Article : Google Scholar : PubMed/NCBI

37 

García-Peydró M, Fuentes P, Mosquera M, García-León MJ, Alcain J, Rodríguez A, García de Miguel P, Menéndez P, Weijer K, Spits H, et al: The NOTCH1/CD44 axis drives pathogenesis in a T cell acute lymphoblastic leukemia model. J Clin Invest. 128:2802–2818. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Rangarajan A, Talora C, Okuyama R, Nicolas M, Mammucari C, Oh H, Aster JC, Krishna S, Metzger D, Chambon P, et al: Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. EMBO J. 20:3427–3436. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Procopio MG, Laszlo C, Al Labban D, Kim DE, Bordignon P, Jo SH, Goruppi S, Menietti E, Ostano P, Ala U, et al: Combined CSL and p53 downregulation promotes cancer-associated fibroblast activation. Nat Cell Biol. 17:1193–1204. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Jarriault S, Le Bail O, Hirsinger E, Pourquié O, Logeat F, Strong CF, Brou C, Seidah NG and Isra l A: Delta-1 activation of Notch-1 signaling results in HES-1 transactivation. Mol Cell Biol. 18:7423–7431. 1998. View Article : Google Scholar : PubMed/NCBI

41 

Lim JS, Ibaseta A, Fischer MM, Cancilla B, O'Young G, Cristea S, Luca VC, Yang D, Jahchan NS, Hamard C, et al: Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature. 545:360–364. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Stoeck A, Lejnine S, Truong A, Pan L, Wang H, Zang C, Yuan J, Ware C, MacLean J, Garrett-Engele PW, et al: Discovery of biomarkers predictive of GSI response in triple-negative breast cancer and adenoid cystic carcinoma. Cancer Discov. 4:1154–1167. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Maier MM and Gessler M: Comparative analysis of the human and mouse Hey1 promoter: Hey genes are new Notch target genes. Biochem Biophys Res Commun. 275:652–660. 2000. View Article : Google Scholar : PubMed/NCBI

44 

Weng AP, Millholland JM, Yashiro-Ohtani Y, Arcangeli ML, Lau A, Wai C, Del Bianco C, Rodriguez CG, Sai H, Tobias J, et al: c-Myc is an important direct target of Notch1 in T-cell acute lymphoblastic leukemia/lymphoma. Genes Dev. 20:2096–2109. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Gekas C, D'Altri T, Aligué R, González J, Espinosa L and Bigas A: β-Catenin is required for T-cell leukemia initiation and MYC transcription downstream of Notch1. Leukemia. 30:2002–2010. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Tottone L, Zhdanovskaya N, Carmona Pestaña Á, Zampieri M, Simeoni F, Lazzari S, Ruocco V, Pelullo M, Caiafa P, Felli MP, et al: Histone modifications drive aberrant Notch3 expression/activity and growth in T-ALL. Front Oncol. 9:1982019. View Article : Google Scholar : PubMed/NCBI

47 

Pirot P, van Grunsven LA, Marine JC, Huylebroeck D and Bellefroid EJ: Direct regulation of the Nrarp gene promoter by the Notch signaling pathway. Biochem Biophys Res Commun. 322:526–534. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Wakabayashi N, Skoko JJ, Chartoumpekis DV, Kimura S, Slocum SL, Noda K, Palliyaguru DL, Fujimuro M, Boley PA, Tanaka Y, et al: Notch-Nrf2 axis: Regulation of Nrf2 gene expression and cytoprotection by Notch signaling. Mol Cell Biol. 34:653–663. 2014. View Article : Google Scholar :

49 

VanDussen KL, Carulli AJ, Keeley TM, Patel SR, Puthoff BJ, Magness ST, Tran IT, Maillard I, Siebel C, Kolterud Å, et al: Notch signaling modulates proliferation and differentiation of intestinal crypt base columnar stem cells. Development. 139:488–497. 2012. View Article : Google Scholar :

50 

Weber BN, Chi AW, Chavez A, Yashiro-Ohtani Y, Yang Q, Shestova O and Bhandoola A: A critical role for TCF-1 in T-lineage specification and differentiation. Nature. 476:63–68. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Germar K, Dose M, Konstantinou T, Zhang J, Wang H, Lobry C, Arnett KL, Blacklow SC, Aifantis I, Aster JC and Gounari F: T-cell factor 1 is a gatekeeper for T-cell specification in response to Notch signaling. Proc Natl Acad Sci USA. 108:20060–20065. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Bray SJ and Gomez-Lamarca M: Notch after cleavage. Curr Opin Cell Biol. 51:103–109. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Chen B, Jiang L, Zhong ML, Li JF, Li BS, Peng LJ, Dai YT, Cui BW, Yan TQ, Zhang WN, et al: Identification of fusion genes and characterization of transcriptome features in T-cell acute lymphoblastic leukemia. Proc Natl Acad Sci USA. 115:373–378. 2018. View Article : Google Scholar

54 

Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, Dimitriadoy S, Liu DL, Kantheti HS, Saghafinia S, et al: Oncogenic signaling pathways in The Cancer Genome Atlas. Cell. 173:321–337.e10. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Weng AP, Ferrando AA, Lee W, Morris JP IV, Silverman LB, Sanchez-Irizarry C, Blacklow SC, Look AT and Aster JC: Activating mutations of NOTCH1 in human T cell acute lympho-blastic leukemia. Science. 306:269–271. 2004. View Article : Google Scholar : PubMed/NCBI

56 

Palomero T, Barnes KC, Real PJ, Glade Bender JL, Sulis ML, Murty VV, Colovai AI, Balbin M and Ferrando AA: CUTLL1, a novel human T-cell lymphoma cell line with t(7;9) rearrangement, aberrant NOTCH1 activation and high sensitivity to gamma-secretase inhibitors. Leukemia. 20:1279–1287. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Bie De J, Demeyer S, Alberti-Servera L, Geerdens E, Segers H, Broux M, De Keersmaecker K, Michaux L, Vandenberghe P, Voet T, et al: Single-cell sequencing reveals the origin and the order of mutation acquisition in T-cell acute lymphoblastic leukemia. Leukemia. 32:1358–1369. 2018. View Article : Google Scholar

58 

Puente XS, Pinyol M, Quesada V, Conde L, Ordóñez GR, Villamor N, Escaramis G, Jares P, Beà S, González-Díaz M, et al: Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature. 475:101–105. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Fabbri G and Dalla-Favera R: The molecular pathogenesis of chronic lymphocytic leukaemia. Nat Rev Cancer. 16:145–162. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Karube K, Enjuanes A, Dlouhy I, Jares P, Martin-Garcia D, Nadeu F, Ordóñez GR, Rovira J, Clot G, Royo C, et al: Integrating genomic alterations in diffuse large B-cell lymphoma identifies new relevant pathways and potential therapeutic targets. Leukemia. 32:675–684. 2018. View Article : Google Scholar :

61 

González-Rincón J, Méndez M, Gómez S, García JF, Martín P, Bellas C, Pedrosa L, Rodríguez-Pinilla SM, Camacho FI, Quero C, et al: Unraveling transformation of follicular lymphoma to diffuse large B-cell lymphoma. PLoS One. 14:e02128132019. View Article : Google Scholar : PubMed/NCBI

62 

Kridel R, Meissner B, Rogic S, Boyle M, Telenius A, Woolcock B, Gunawardana J, Jenkins C, Cochrane C, Ben-Neriah S, et al: Whole transcriptome sequencing reveals recurrent NOTCH1 mutations in mantle cell lymphoma. Blood. 119:1963–1971. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Robinson DR, Kalyana-Sundaram S, Wu YM, Shankar S, Cao X, Ateeq B, Asangani IA, Iyer M, Maher CA, Grasso CS, et al: Functionally recurrent rearrangements of the MAST kinase and Notch gene families in breast cancer. Nat Med. 17:1646–1651. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Wang K, Zhang Q, Li D, Ching K, Zhang C, Zheng X, Ozeck M, Shi S, Li X, Wang H, et al: PEST domain mutations in Notch receptors comprise an oncogenic driver segment in triple-negative breast cancer sensitive to a γ-secretase inhibitor. Clin Cancer Res. 21:1487–1496. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Robinson DR, Wu YM, Lonigro RJ, Vats P, Cobain E, Everett J, Cao X, Rabban E, Kumar-Sinha C, Raymond V, et al: Integrative clinical genomics of metastatic cancer. Nature. 548:297–303. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Westhoff B, Colaluca IN, D'Ario G, Donzelli M, Tosoni D, Volorio S, Pelosi G, Spaggiari L, Mazzarol G, Viale G, et al: Alterations of the Notch pathway in lung cancer. Proc Natl Acad Sci USA. 106:22293–22298. 2009. View Article : Google Scholar : PubMed/NCBI

67 

Wang NJ, Sanborn Z, Arnett KL, Bayston LJ, Liao W, Proby CM, Leigh IM, Collisson EA, Gordon PB, Jakkula L, et al: Loss-of-function mutations in Notch receptors in cutaneous and lung squamous cell carcinoma. Proc Natl Acad Sci USA. 108:17761–17766. 2011. View Article : Google Scholar : PubMed/NCBI

68 

Agrawal N, Frederick MJ, Pickering CR, Bettegowda C, Chang K, Li RJ, Fakhry C, Xie TX, Zhang J, Wang J, et al: Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science. 333:1154–1157. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Stransky N, Egloff AM, Tward AD, Kostic AD, Cibulskis K, Sivachenko A, Kryukov GV, Lawrence MS, Sougnez C, McKenna A, et al: The mutational landscape of head and neck squamous cell carcinoma. Science. 333:1157–1160. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Song Y, Li L, Ou Y, Gao Z, Li E, Li X, Zhang W, Wang J, Xu L, Zhou Y, et al: Identification of genomic alterations in oesophageal squamous cell cancer. Nature. 509:91–95. 2014. View Article : Google Scholar : PubMed/NCBI

71 

Martincorena I, Fowler JC, Wabik A, Lawson ARJ, Abascal F, Hall MWJ, Cagan A, Murai K, Mahbubani K, Stratton MR, et al: Somatic mutant clones colonize the human esophagus with age. Science. 362:911–917. 2018. View Article : Google Scholar : PubMed/NCBI

72 

George J, Lim JS, Jang SJ, Cun Y, Ozretić L, Kong G, Leenders F, Lu X, Fernández-Cuesta L, Bosco G, et al: Comprehensive genomic profiles of small cell lung cancer. Nature. 524:47–53. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Ali SA, Justilien V, Jamieson L, Murray NR and Fields AP: Protein kinase Cι drives a NOTCH3-dependent stem-like phenotype in mutant KRAS lung adenocarcinoma. Cancer Cell. 29:367–378. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Bhagat TD, Zou Y, Huang S, Park J, Palmer MB, Hu C, Li W, Shenoy N, Giricz O, Choudhary G, et al: Notch pathway is activated via genetic and epigenetic alterations and is a therapeutic target in clear cell renal cancer. J Biol Chem. 292:837–846. 2017. View Article : Google Scholar :

75 

van Groningen T, Akogul N, Westerhout EM, Chan A, Hasselt NE, Zwijnenburg DA, Broekmans M, Stroeken P, Haneveld F, Hooijer GKJ, et al: A NOTCH feed-forward loop drives reprogramming from adrenergic to mesenchymal state in neuroblastoma. Nat Commun. 10:15302019. View Article : Google Scholar : PubMed/NCBI

76 

Ban J, Bennani-Baiti IM, Kauer M, Schaefer KL, Poremba C, Jug G, Schwentner R, Smrzka O, Muehlbacher K, Aryee DN and Kovar H: EWS-FLI1 suppresses NOTCH-activated p53 in Ewing's sarcoma. Cancer Res. 68:7100–7109. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Augert A, Eastwood E, Ibrahim AH, Wu N, Grunblatt E, Basom R, Liggitt D, Eaton KD, Martins R, Poirier JT, et al: Targeting NOTCH activation in small cell lung cancer through LSD1 inhibition. Sci Signal. 12:pii: eaau2922. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Song Y, Zhang Y, Jiang H, Zhu Y, Liu L, Feng W, Yang L, Wang Y and Li M: Activation of Notch3 promotes pulmonary arterial smooth muscle cells proliferation via Hes1/p27Kip1 signaling pathway. FEBS Open Bio. 5:656–660. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Maraver A, Fernández-Marcos PJ, Herranz D, Muñoz-Martin M, Gomez-Lopez G, Cañamero M, Mulero F, Megías D, Sanchez-Carbayo M, Shen J, et al: Therapeutic effect of γ-secretase inhibition in KrasG12V-driven non-small cell lung carcinoma by derepression of DUSP1 and inhibition of ERK. Cancer Cell. 22:222–234. 2012. View Article : Google Scholar : PubMed/NCBI

80 

Palomero T, Sulis ML, Cortina M, Real PJ, Barnes K, Ciofani M, Caparros E, Buteau J, Brown K, Perkins SL, et al: Mutational loss of PTEN induces resistance to NOTCH1 inhibition in T-cell leukemia. Nat Med. 13:1203–1210. 2007. View Article : Google Scholar : PubMed/NCBI

81 

Zhou X, Smith AJ, Waterhouse A, Blin G, Malaguti M, Lin CY, Osorno R, Chambers I and Lowell S: Hes1 desynchronizes differentiation of pluripotent cells by modulating STAT3 activity. Stem Cells. 31:1511–1122. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Weng MT, Tsao PN, Lin HL, Tung CC, Change MC, Chang YT, Wong JM and Wei SC: Hes1 increases the invasion ability of colorectal cancer cells via the STAT3-MMP14 pathway. PLoS One. 10:e01443222015. View Article : Google Scholar : PubMed/NCBI

83 

Jin S, Mutvei AP, Chivukula IV, Andersson ER, Ramsköld D, Sandberg R, Lee KL, Kronqvist P, Mamaeva V, Ostling P, et al: Non-canonical Notch signaling activates IL-6/JAK/STAT signaling in breast tumor cells and is controlled by p53 and IKKα/IKKβ. Oncogene. 32:4892–4902. 2013. View Article : Google Scholar

84 

Schreck KC, Taylor P, Marchionni L, Gopalakrishnan V, Bar EE, Gaiano N and Eberhart CG: The Notch target Hes1 directly modulates Gli1 expression and Hedgehog signaling: A potential mechanism of therapeutic resistance. Clin Cancer Res. 16:6060–6070. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Bonyadi Rad E, Hammerlindl H, Wels C, Popper U, Ravindran Menon D, Breiteneder H, Kitzwoegerer M, Hafner C, Herlyn M, Bergler H and Schaider H: Notch4 signaling induces a mesenchymal-epithelial-like transition in melanoma cells to suppress malignant behaviors. Cancer Res. 76:1690–1697. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Wang D, Xu J, Liu B, He X, Zhou L, Hu X, Qiao F, Zhang A, Xu X, Zhang H, et al: IL6 blockade potentiates the anti-tumor effects of γ-secretase inhibitors in Notch3-expressing breast cancer. Cell Death Differ. 25:330–339. 2018. View Article : Google Scholar

87 

Hartman BH, Reh TA and Bermingham-McDonogh O: Notch signaling specifies prosensory domains via lateral induction in the developing mammalian inner ear. Proc Natl Acad Sci USA. 107:15792–15797. 2010. View Article : Google Scholar : PubMed/NCBI

88 

Petrovic J, Formosa-Jordan P, Luna-Escalante JC, Abelló G, Ibañes M, Neves J and Giraldez F: Ligand-dependent Notch signaling strength orchestrates lateral induction and lateral inhibition in the developing inner ear. Development. 141:2313–2324. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Sun W, Gaykalova DA, Ochs MF, Mambo E, Arnaoutakis D, Liu Y, Loyo M, Agrawal N, Howard J, Li R, et al: Activation of the NOTCH pathway in head and neck cancer. Cancer Res. 74:1091–1104. 2014. View Article : Google Scholar :

90 

Turley SJ, Cremasco V and Astarita JL: Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol. 15:669–682. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Valkenburg KC, de Groot AE and Pienta KJ: Targeting the tumour stroma to improve cancer therapy. Nat Rev Clin Oncol. 15:366–381. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Östman A and Corvigno S: Microvascular mural cells in cancer. Trends Cancer. 4:838–848. 2018. View Article : Google Scholar : PubMed/NCBI

93 

Lambrechts D, Wauters E, Boeckx B, Aibar S, Nittner D, Burton O, Bassez A, Decaluwé H, Pircher A, Van den Eynde K, et al: Phenotype molding of stromal cells in the lung tumor microenvironment. Nat Med. 24:1277–1289. 2018. View Article : Google Scholar : PubMed/NCBI

94 

Bartoschek M, Oskolkov N, Bocci M, Lövrot J, Larsson C, Sommarin M, Madsen CD, Lindgren D, Pekar G, Karlsson G, et al: Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing. Nat Commun. 9:51502018. View Article : Google Scholar : PubMed/NCBI

95 

Kalluri R: The biology and function of fibroblasts in cancer. Nat Rev Cancer. 16:582–598. 2016. View Article : Google Scholar : PubMed/NCBI

96 

Altorki NK, Markowitz GJ, Gao D, Port JL, Saxena A, Stiles B, McGraw T and Mittal V: The lung microenvironment: An important regulator of tumour growth and metastasis. Nat Rev Cancer. 19:9–31. 2019. View Article : Google Scholar :

97 

Wang Z and Zöller M: Exosomes, metastases, and the miracle of cancer stem cell markers. Cancer Metastasis Rev. 38:259–295. 2019. View Article : Google Scholar : PubMed/NCBI

98 

Katoh M: Canonical and non-canonical WNT signaling in cancer stem cells and their niches: Cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review). Int J Oncol. 51:1357–1369. 2017. View Article : Google Scholar : PubMed/NCBI

99 

Dotto GP: Multifocal epithelial tumors and field cancerization: Stroma as a primary determinant. J Clin Invest. 124:1446–1453. 2014. View Article : Google Scholar : PubMed/NCBI

100 

Shibue T and Weinberg RA: EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat Rev Clin Oncol. 14:611–629. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Schreiber RD, Old LJ and Smyth MJ: Cancer immunoediting: Integrating immunity's roles in cancer suppression and promotion. Science. 331:1565–1570. 2011. View Article : Google Scholar : PubMed/NCBI

102 

Fukumura D, Kloepper J, Amoozgar Z, Duda DG and Jain RK: Enhancing cancer immunotherapy using antiangiogenics: Opportunities and challenges. Nat Rev Clin Oncol. 15:325–340. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Robbins J, Blondel BJ, Gallahan D and Callahan R: Mouse mammary tumor gene int-3: A member of the Notch gene family transforms mammary epithelial cells. J Virol. 66:2594–2599. 1992. View Article : Google Scholar : PubMed/NCBI

104 

Peters G, Lee AE and Dickson C: Concerted activation of two potential proto-oncogenes in carcinomas induced by mouse mammary tumour virus. Nature. 320:628–631. 1986. View Article : Google Scholar : PubMed/NCBI

105 

Shackleford GM, MacArthur CA, Kwan HC and Varmus HE: Mouse mammary tumor virus infection accelerates mammary carcinogenesis in Wnt-1 transgenic mice by insertional activation of int-2/Fgf-3 and hst/Fgf-4. Proc Natl Acad Sci USA. 90:740–744. 1993. View Article : Google Scholar : PubMed/NCBI

106 

Katoh M: WNT and FGF gene clusters (review). Int J Oncol. 21:1269–1273. 2002.PubMed/NCBI

107 

Lowther W, Wiley K, Smith GH and Callahan R: A new common integration site, Int7, for the mouse mammary tumor virus in mouse mammary tumors identifies a gene whose product has furin-like and thrombospondin-like sequences. J Virol. 79:10093–10096. 2005. View Article : Google Scholar : PubMed/NCBI

108 

Theodorou V, Kimm MA, Boer M, Wessels L, Theelen W, Jonkers J and Hilkens J: MMTV insertional mutagenesis identifies genes, gene families and pathways involved in mammary cancer. Nat Genet. 39:759–769. 2007. View Article : Google Scholar : PubMed/NCBI

109 

Zhan T, Rindtorff N and Boutros M: Wnt signaling in cancer. Oncogene. 36:1461–1473. 2017. View Article : Google Scholar :

110 

Morgan RG, Mortensson E and Williams AC: Targeting LGR5 in colorectal cancer: Therapeutic gold or too plastic. Br J Cancer. 118:1410–1418. 2018. View Article : Google Scholar : PubMed/NCBI

111 

Estrach S, Ambler CA, Lo Celso C, Hozumi K and Watt FM: Jagged 1 is a beta-catenin target gene required for ectopic hair follicle formation in adult epidermis. Development. 133:4427–4438. 2006. View Article : Google Scholar : PubMed/NCBI

112 

Ma L, Wang Y, Hui Y, Du Y, Chen Z, Feng H, Zhang S, Li N, Song J, Fang Y, et al: WNT/NOTCH pathway is essential for the maintenance and expansion of human MGE progenitors. Stem Cell Reports. 12:934–949. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Boulter L, Govaere O, Bird TG, Radulescu S, Ramachandran P, Pellicoro A, Ridgway RA, Seo SS, Spee B, Van Rooijen N, et al: Macrophage-derived Wnt opposes Notch signaling to specify hepatic progenitor cell fate in chronic liver disease. Nat Med. 18:572–579. 2012. View Article : Google Scholar : PubMed/NCBI

114 

Högström J, Heino S, Kallio P, Lähde M, Leppänen VM, Balboa D, Wiener Z and Alitalo K: Transcription factor PROX1 suppresses Notch pathway activation via the nucleosome remod-eling and deacetylase complex in colorectal cancer stem-like cells. Cancer Res. 78:5820–5832. 2018.

115 

Phng LK, Potente M, Leslie JD, Babbage J, Nyqvist D, Lobov I, Ondr JK, Rao S, Lang RA, Thurston G and Gerhardt H: Nrarp coordinates endothelial Notch and Wnt signaling to control vessel density in angiogenesis. Dev Cell. 16:70–82. 2009. View Article : Google Scholar : PubMed/NCBI

116 

Liu W, Li H, Hong SH, Piszczek GP, Chen W and Rodgers GP: Olfactomedin 4 deletion induces colon adenocarcinoma in ApcMin/+ mice. Oncogene. 35:5237–5247. 2016. View Article : Google Scholar : PubMed/NCBI

117 

Giancotti FG: Mechanisms governing metastatic dormancy and reactivation. Cell. 155:750–764. 2013. View Article : Google Scholar : PubMed/NCBI

118 

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

119 

Dimri GP, Martinez JL, Jacobs JJ, Keblusek P, Itahana K, Van Lohuizen M, Campisi J, Wazer DE and Band V: The Bmi-1 oncogene induces telomerase activity and immortalizes human mammary epithelial cells. Cancer Res. 62:4736–4745. 2002.PubMed/NCBI

120 

De Jaime-Soguero A, Aulicino F, Ertaylan G, Griego A, Cerrato A, Tallam A, Del Sol A, Cosma MP and Lluis F: Wnt/Tcf1 pathway restricts embryonic stem cell cycle through activation of the Ink4/Arf locus. PLoS Genet. 13:e10066822017. View Article : Google Scholar : PubMed/NCBI

121 

Srinivasan T, Walters J, Bu P, Than EB, Tung KL, Chen KY, Panarelli N, Milsom J, Augenlicht L, Lipkin SM and Shen X: NOTCH signaling regulates asymmetric cell fate of fast- and slow-cycling colon cancer-initiating cells. Cancer Res. 76:3411–3421. 2016. View Article : Google Scholar : PubMed/NCBI

122 

Germann M, Xu H, Malaterre J, Sampurno S, Huyghe M, Cheasley D, Fre S and Ramsay RG: Tripartite interactions between Wnt signaling, Notch and Myb for stem/progenitor cell functions during intestinal tumorigenesis. Stem Cell Res. 13:355–366. 2014. View Article : Google Scholar : PubMed/NCBI

123 

Mourao L, Jacquemin G, Huyghe M, Nawrocki WJ, Menssouri N, Servant N and Fre S: Lineage tracing of Notch1-expressing cells in intestinal tumours reveals a distinct population of cancer stem cells. Sci Rep. 9:8882019. View Article : Google Scholar : PubMed/NCBI

124 

Jain RK: Normalizing tumor microenvironment to treat cancer: Bench to bedside to biomarkers. J Clin Oncol. 31:2205–2218. 2013. View Article : Google Scholar : PubMed/NCBI

125 

De Palma M, Biziato D and Petrova TV: Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer. 17:457–474. 2017. View Article : Google Scholar : PubMed/NCBI

126 

Yunus M, Jansson PJ, Kovacevic Z, Kalinowski DS and Richardson DR: Tumor-induced neoangiogenesis and receptor tyrosine kinases-Mechanisms and strategies for acquired resistance. Biochim Biophys Acta Gen Subj. 1863:1217–1225. 2019. View Article : Google Scholar : PubMed/NCBI

127 

Potente M, Gerhardt H and Carmeliet P: Basic and therapeutic aspects of angiogenesis. Cell. 146:873–887. 2011. View Article : Google Scholar : PubMed/NCBI

128 

Goel HL and Mercurio AM: VEGF targets the tumour cell. Nat Rev Cancer. 13:871–882. 2013. View Article : Google Scholar : PubMed/NCBI

129 

Bridges E, Oon CE and Harris A: Notch regulation of tumor angiogenesis. Future Oncol. 7:569–588. 2011. View Article : Google Scholar : PubMed/NCBI

130 

Simons M, Gordon E and Claesson-Welsh L: Mechanisms and regulation of endothelial VEGF receptor signalling. Nat Rev Mol Cell Biol. 17:611–625. 2016. View Article : Google Scholar : PubMed/NCBI

131 

Wieland E, Rodriguez-Vita J, Liebler SS, Mogler C, Moll I, Herberich SE, Espinet E, Herpel E, Menuchin A, Chang-Claude J, et al: Endothelial Notch1 activity facilitates metastasis. Cancer Cell. 31:355–367. 2017. View Article : Google Scholar : PubMed/NCBI

132 

Wohlfeil SA, Häfele V, Dietsch B, Schledzewski K, Winkler M, Zierow J, Leibing T, Mohammadi MM, Heineke J, Sticht C, et al: Hepatic endothelial Notch activation protects against liver metastasis by regulating endothelial-tumor cell adhesion independent of angiocrine signaling. Cancer Res. 79:598–610. 2019. View Article : Google Scholar

133 

Radtke F, MacDonald HR and Tacchini-Cottier F: Regulation of innate and adaptive immunity by Notch. Nat Rev Immunol. 13:427–437. 2013. View Article : Google Scholar : PubMed/NCBI

134 

Lobry C, Oh P, Mansour MR, Look AT and Aifantis I: Notch signaling: Switching an oncogene to a tumor suppressor. Blood. 123:2451–2459. 2014. View Article : Google Scholar : PubMed/NCBI

135 

Amsen D, Helbig C and Backer RA: Notch in T cell differentiation: all things considered. Trends Immunol. 36:802–814. 2015. View Article : Google Scholar : PubMed/NCBI

136 

Charbonnier LM, Wang S, Georgiev P, Sefik E and Chatila TA: Control of peripheral tolerance by regulatory T cell-intrinsic Notch signaling. Nat Immunol. 16:1162–1173. 2015. View Article : Google Scholar : PubMed/NCBI

137 

Wang YC, He F, Feng F, Liu XW, Dong GY, Qin HY, Hu XB, Zheng MH, Liang L, Feng L, et al: Notch signaling determines the M1 versus M2 polarization of macrophages in antitumor immune responses. Cancer Res. 70:4840–4849. 2010. View Article : Google Scholar : PubMed/NCBI

138 

Liu H, Wang J, Zhang M, Xuan Q, Wang Z, Lian X and Zhang Q: Jagged1 promotes aromatase inhibitor resistance by modulating tumor-associated macrophage differentiation in breast cancer patients. Breast Cancer Res Treat. 166:95–107. 2017. View Article : Google Scholar : PubMed/NCBI

139 

Rashedi I, Gómez-Aristizábal A, Wang XH, Viswanathan S and Keating A: TLR3 or TLR4 activation enhances mesenchymal stromal cell-mediated Treg induction via Notch signaling. Stem Cells. 35:265–275. 2017. View Article : Google Scholar

140 

Cahill EF, Tobin LM, Carty F, Mahon BP and English K: Jagged-1 is required for the expansion of CD4+ CD25+ FoxP3+ regulatory T cells and tolerogenic dendritic cells by murine mesenchymal stromal cells. Stem Cell Res Ther. 6:192015. View Article : Google Scholar : PubMed/NCBI

141 

Kared H, Adle-Biassette H, Foïs E, Masson A, Bach JF, Chatenoud L, Schneider E and Zavala F: Jagged2-expressing hematopoietic progenitors promote regulatory T cell expansion in the periphery through Notch signaling. Immunity. 25:823–834. 2006. View Article : Google Scholar : PubMed/NCBI

142 

Ting HA, de Almeida Nagata D, Rasky AJ, Malinczak CA, Maillard IP, Schaller MA and Lukacs NW: Notch ligand Delta-like 4 induces epigenetic regulation of Treg cell differentiation and function in viral infection. Mucosal Immunol. 11:1524–1536. 2018. View Article : Google Scholar : PubMed/NCBI

143 

Zhang L, Yu X, Zheng L, Zhang Y, Li Y, Fang Q, Gao R, Kang B, Zhang Q, Huang JY, et al: Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature. 564:268–272. 2018. View Article : Google Scholar : PubMed/NCBI

144 

Gustafsson MV, Zheng X, Pereira T, Gradin K, Jin S, Lundkvist J, Ruas JL, Poellinger L, Lendahl U and Bondesson M: Hypoxia requires Notch signaling to maintain the undifferentiated cell state. Dev Cell. 9:617–628. 2005. View Article : Google Scholar : PubMed/NCBI

145 

Katoh M: FGFR inhibitors: Effects on cancer cells, tumor microenvironment and whole-body homeostasis (Review). Int J Mol Med. 38:3–15. 2016. View Article : Google Scholar : PubMed/NCBI

146 

Grazioli P, Felli MP, Screpanti I and Campese AF: The mazy case of Notch and immunoregulatory cells. J Leukoc Biol. 102:361–368. 2017. View Article : Google Scholar : PubMed/NCBI

147 

Katoh M: Genomic testing, tumor microenvironment and targeted therapy of Hedgehog-related human cancers. Clin Sci (Lond). 133:953–970. 2019. View Article : Google Scholar

148 

Yang Z, Qi Y, Lai N, Zhang J, Chen Z, Liu M, Zhang W, Luo R and Kang S: Notch1 signaling in melanoma cells promoted tumor-induced immunosuppression via upregulation of TGF-β1. J Exp Clin Cancer Res. 37:12018. View Article : Google Scholar

149 

Mao L, Zhao ZL, Yu GT, Wu L, Deng WW, Li YC, Liu JF, Bu LL, Liu B, Kulkarni AB, et al: γ-Secretase inhibitor reduces immunosuppressive cells and enhances tumour immunity in head and neck squamous cell carcinoma. Int J Cancer. 142:999–1009. 2018. View Article : Google Scholar

150 

El-Khoueiry AB, Desai J, Iyer SP, Gadgeel SM, Ramalingam SS, Horn L, LoRusso P, Bajaj G, Kollia G, Qi Z, et al: A phase I study of AL101, a pan-NOTCH inhibitor, in patients (pts) with locally advanced or metastatic solid tumors. J Clin Oncol. 36(15 Suppl): S25152018. View Article : Google Scholar

151 

Massard C, Azaro A, Soria JC, Lassen U, Le Tourneau C, Sarker D, Smith C, Ohnmacht U, Oakley G, Patel BKR, et al: First-in-human study of LY3039478, an oral Notch signaling inhibitor in advanced or metastatic cancer. Ann Oncol. 29:1911–1917. 2018. View Article : Google Scholar : PubMed/NCBI

152 

Habets RA, de Bock CE, Serneels L, Lodewijckx I, Verbeke D, Nittner D, Narlawar R, Demeyer S, Dooley J, Liston A, et al: Safe targeting of T cell acute lymphoblastic leukemia by pathology-specific NOTCH inhibition. Sci Transl Med. 11:pii: eaau6246. 2019. View Article : Google Scholar : PubMed/NCBI

153 

Messersmith WA, Shapiro GI, Cleary JM, Jimeno A, Dasari A, Huang B, Shaik MN, Cesari R, Zheng X, Reynolds JM, et al: A phase I, dose-finding study in patients with advanced solid malignancies of the oral γ-secretase inhibitor PF-03084014. Clin Cancer Res. 21:60–67. 2015. View Article : Google Scholar

154 

Kummar S, O'Sullivan Coyne G, Do KT, Turkbey B, Meltzer PS, Polley E, Choyke PL, Meehan R, Vilimas R, Horneffer Y, et al: Clinical activity of the γ-secretase inhibitor PF-03084014 in adults with desmoid tumors (aggressive fibromatosis). J Clin Oncol. 35:1561–1569. 2017. View Article : Google Scholar : PubMed/NCBI

155 

Tolcher AW, Messersmith WA, Mikulski SM, Papadopoulos KP, Kwak EL, Gibbon DG, Patnaik A, Falchook GS, Dasari A, Shapiro GI, et al: Phase I study of RO4929097, a gamma secre-tase inhibitor of Notch signaling, in patients with refractory metastatic or locally advanced solid tumors. J Clin Oncol. 30:2348–2353. 2012. View Article : Google Scholar : PubMed/NCBI

156 

Strosberg JR, Yeatman T, Weber J, Coppola D, Schell MJ, Han G, Almhanna K, Kim R, Valone T, Jump H and Sullivan D: A phase II study of RO4929097 in metastatic colorectal cancer. Eur J Cancer. 48:997–1003. 2012. View Article : Google Scholar : PubMed/NCBI

157 

Jordan NV, Bardia A, Wittner BS, Benes C, Ligorio M, Zheng Y, Yu M, Sundaresan TK, Licausi JA, Desai R, et al: HER2 expression identifies dynamic functional states within circulating breast cancer cells. Nature. 537:102–106. 2016. View Article : Google Scholar : PubMed/NCBI

158 

Diluvio G, Del Gaudio F, Giuli MV, Franciosa G, Giuliani E, Palermo R, Besharat ZM, Pignataro MG, Vacca A, d'Amati G, et al: NOTCH3 inactivation increases triple negative breast cancer sensitivity to gefitinib by promoting EGFR tyrosine dephosphorylation and its intracellular arrest. Oncogenesis. 7:422018. View Article : Google Scholar : PubMed/NCBI

159 

Yao J, Qian C, Shu T, Zhang X, Zhao Z and Liang Y: Combination treatment of PD98059 and DAPT in gastric cancer through induction of apoptosis and downregulation of WNT/β-catenin. Cancer Biol Ther. 14:833–839. 2013. View Article : Google Scholar : PubMed/NCBI

160 

Smith DC, Eisenberg PD, Manikhas G, Chugh R, Gubens MA, Stagg RJ, Kapoun AM, Xu L, Dupont J and Sikic B: A phase I dose escalation and expansion study of the anticancer stem cell agent demcizumab (anti-DLL4) in patients with previously treated solid tumors. Clin Cancer Res. 20:6295–6303. 2014. View Article : Google Scholar : PubMed/NCBI

161 

Chiorean EG, LoRusso P, Strother RM, Diamond JR, Younger A, Messersmith WA, Adriaens L, Liu L, Kao RJ, DiCioccio AT, et al: A phase I first-in-human study of enoticumab (REGN421), a fully human Delta-like ligand 4 (DLL4) monoclonal antibody, in patients with advanced solid tumors. Clin Cancer Res. 21:2695–2703. 2015. View Article : Google Scholar : PubMed/NCBI

162 

Falchook GS, Dowlati A, Naing A, Gribbin MJ, Jenkins DW, Chang LL, Lai DW and Smith DC: Phase I study of MEDI0639 in patients with advanced solid tumors. J Clin Oncol. 33(15 Suppl): S30242015. View Article : Google Scholar

163 

Ferrarotto R, Eckhardt G, Patnaik A, LoRusso P, Faoro L, Heymach JV, Kapoun AM, Xu L and Munster P: A phase I dose-escalation and dose-expansion study of brontictuzumab in subjects with selected solid tumors. Ann Oncol. 29:1561–1568. 2018. View Article : Google Scholar : PubMed/NCBI

164 

Yen WC, Fischer MM, Axelrod F, Bond C, Cain J, Cancilla B, Henner WR, Meisner R, Sato A, Shah J, et al: Targeting Notch signaling with a Notch2/Notch3 antagonist (tarextumab) inhibits tumor growth and decreases tumor-initiating cell frequency. Clin Cancer Res. 21:2084–2095. 2015. View Article : Google Scholar : PubMed/NCBI

165 

Smith DC, Chugh R, Patnaik A, Papadopoulos KP, Wang M, Kapoun AM, Xu L, Dupont J, Stagg RJ and Tolcher A: A phase 1 dose escalation and expansion study of Tarextumab (OMP-59R5) in patients with solid tumors. Invest New Drugs. 37:722–730. 2019. View Article : Google Scholar :

166 

Beck A, Goetsch L, Dumontet C and Corvaïa N: Strategies and challenges for the next generation of antibody-drug conjugates. Nat Rev Drug Discov. 16:315–337. 2017. View Article : Google Scholar : PubMed/NCBI

167 

Lambert JM and Berkenblit A: Antibody-Drug conjugates for cancer treatment. Annu Rev Med. 69:191–207. 2018. View Article : Google Scholar : PubMed/NCBI

168 

Carter PJ and Lazar GA: Next generation antibody drugs: Pursuit of the 'high-hanging fruit'. Nat Rev Drug Discov. 17:197–223. 2018. View Article : Google Scholar

169 

June CH, O'Connor RS, Kawalekar OU, Ghassemi S and Milone MC: CAR T cell immunotherapy for human cancer. Science. 359:1361–1365. 2018. View Article : Google Scholar : PubMed/NCBI

170 

Saunders LR, Bankovich AJ, Anderson WC, Aujay MA, Bheddah S, Black K, Desai R, Escarpe PA, Hampl J, Laysang A, et al: A DLL3-targeted antibody-drug conjugate eradicates high-grade pulmonary neuroendocrine tumor-initiating cells in vivo. Sci Transl Med. 7:302ra1362015. View Article : Google Scholar : PubMed/NCBI

171 

Rudin CM, Pietanza MC, Bauer TM, Ready N, Morgensztern D, Glisson BS, Byers LA, Johnson ML, Burris HA III, Robert F, et al: Rovalpituzumab tesirine, a DLL3-targeted antibody-drug conjugate, in recurrent small-cell lung cancer: A first-in-human, first-in-class, open-label, phase 1 study. Lancet Oncol. 18:42–51. 2017. View Article : Google Scholar :

172 

Carbone DP, Morgensztern D, Le Moulec S, Santana-Davila R, Ready N, Hann CL, Glisson BS, Dowlati A, Rudin CM, Lally S, et al: Efficacy and safety of rovalpituzumab tesirine in patients With DLL3-expressing, ≥ 3rd line small cell lung cancer: Results from the phase 2 TRINITY study. J Clin Oncol. 36(15 Suppl): S85072018. View Article : Google Scholar

173 

Rosen LS, Wesolowski R, Baffa R, Liao KH, Hua SY, Gibson BL, Pirie-Shepherd S and Tolcher AW: A phase I, dose-escalation study of PF-06650808, an anti-Notch3 antibody-drug conjugate, in patients with breast cancer and other advanced solid tumors. Invest New Drugs. Mar 18–2019.Epub ahead of print. PubMed/NCBI

174 

Smit MAD, Borghaei H, TOwonikoko TK, Hummel HD, Johnson ML, Champiat S, Salgia R, Udagawa H, Boyer MJ and Govindan R: Phase 1 study of AMG 757, a half-life extended bispecific T cell engager (BiTE) antibody construct targeting DLL3, in patients with small cell lung cancer (SCLC). J Clin Oncol. 37(15 Suppl): TPS85772019.

175 

Li Y, Hickson JA, Ambrosi DJ, Haasch DL, Foster-Duke KD, Eaton LJ, DiGiammarino EL, Panchal SC, Jiang F, Mudd SR, et al: ABT-165, a dual variable domain immunoglobulin (DVD-Ig) targeting DLL4 and VEGF, demonstrates superior efficacy and favorable safety profiles in preclinical models. Mol Cancer Ther. 17:1039–1050. 2018. View Article : Google Scholar : PubMed/NCBI

176 

Jimeno A, Moore KN, Gordon M, Chugh R, Diamond JR, Aljumaily R, Mendelson D, Kapoun AM, Xu L, Stagg R and Smith DC: A first-in-human phase 1a study of the bispecific anti-DLL4/anti-VEGF antibody navicixizumab (OMP-305B83) in patients with previously treated solid tumors. Invest New Drugs. 37:461–472. 2019. View Article : Google Scholar

177 

Hu S, Fu W, Li T, Yuan Q, Wang F, Lv G, Lv Y, Fan X, Shen Y, Lin F, et al: Antagonism of EGFR and Notch limits resistance to EGFR inhibitors and radiation by decreasing tumor-initiating cell frequency. Sci Transl Med. 9:pii: eaag0339. 2017. View Article : Google Scholar

178 

Fu W, Lei C, Yu Y, Liu S, Li T, Lin F, Fan X, Shen Y, Ding M, Tang Y, et al: EGFR/Notch antagonists enhance the response to inhibitors of the PI3K-Akt pathway by decreasing tumor-initiating cell frequency. Clin Cancer Res. 25:2835–2847. 2019. View Article : Google Scholar : PubMed/NCBI

179 

Byers LA, Chiappori A and Smit MAD: Phase 1 study of AMG 119, a chimeric antigen receptor (CAR) T cell therapy targeting DLL3, in patients with relapsed/refractory small cell lung cancer (SCLC). J Clin Oncol. 37(15 Suppl): TPS85762019.

180 

Puca L, Gavyert K, Sailer V, Conteduca V, Dardenne E, Sigouros M, Isse K, Kearney M, Vosoughi A, Fernandez L, et al: Delta-like protein 3 expression and therapeutic targeting in neuroendocrine prostate cancer. Sci Transl Med. 11:pii: eaav0891. 2019. View Article : Google Scholar : PubMed/NCBI

181 

Locke FL, Ghobadi A, Jacobson CA, Miklos DB, Lekakis LJ, Oluwole OO, Lin Y, Braunschweig I, Hill BT, Timmerman JM, et al: Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): A single-arm, multicentre, phase 12 trial. Lancet Oncol. 20:31–42. 2019. View Article : Google Scholar

182 

Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, McGuirk JP, Jäger U, Jaglowski S, Andreadis C, Westin JR, et al: Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 380:45–56. 2019. View Article : Google Scholar

183 

Kantarjian HM, DeAngelo DJ, Stelljes M, Martinelli G, Liedtke M, Stock W, Gökbuget N, O'Brien S, Wang K, Wang T, et al: Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N Engl J Med. 375:740–753. 2016. View Article : Google Scholar : PubMed/NCBI

184 

Horwitz S, O'Connor OA, Pro B, Illidge T, Fanale M, Advani R, Bartlett NL, Christensen JH, Morschhauser F, Domingo-Domenech E, et al: Brentuximab vedotin with chemotherapy for CD30-positive peripheral T-cell lymphoma (ECHELON-2): A global, double-blind, randomised, phase 3 trial. Lancet. 393:229–240. 2019. View Article : Google Scholar :

185 

Tilly H, Morschhauser F, Bartlett NL, Mehta A, Salles G, Haioun C, Munoz J, Chen AI, Kolibaba K, Lu D, et al: Polatuzumab vedotin in combination with immunochemotherapy in patients with previously untreated diffuse large B-cell lymphoma: An open-label, non-randomised, phase 1b-2 study. Lancet Oncol. 20:998–1010. 2019. View Article : Google Scholar : PubMed/NCBI

186 

Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, Pegram M, Oh DY, Diéras V, Guardino E, et al: Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 367:1783–1791. 2012. View Article : Google Scholar : PubMed/NCBI

187 

Doi T, Shitara K, Naito Y, Shimomura A, Fujiwara Y, Yonemori K, Shimizu C, Shimoi T, Kuboki Y, Matsubara N, et al: Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS-8201), a HER2-targeting antibody-drug conjugate, in patients with advanced breast and gastric or gastro-oesophageal tumours: A phase 1 dose-escalation study. Lancet Oncol. 18:1512–1522. 2017. View Article : Google Scholar : PubMed/NCBI

188 

Banerji U, van Herpen CML, Saura C, Thistlethwaite F, Lord S, Moreno V, Macpherson IR, Boni V, Rolfo C, de Vries EGE, et al: Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: A phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 20:1124–1135. 2019. View Article : Google Scholar : PubMed/NCBI

189 

Lassman AB, van den Bent MJ, Gan HK, Reardon DA, Kumthekar P, Butowski N, Lwin Z, Mikkelsen T, Nabors LB, Papadopoulos KP, et al: Safety and efficacy of depatuxizumab mafodotin + temozolomide in patients with EGFR-amplified, recurrent glioblastoma: Results from an international phase I multicenter trial. Neuro Oncol. 21:106–114. 2019. View Article : Google Scholar

190 

Moore KN, Martin LP, O'Malley DM, Matulonis UA, Konner JA, Perez RP, Bauer TM, Ruiz-Soto R and Birrer MJ: Safety and activity of mirvetuximab soravtansine (IMGN853), a folate receptor alpha-targeting antibody-drug conjugate, in platinum-resistant ovarian, fallopian tube, or primary peritoneal cancer: A phase I expansion study. J Clin Oncol. 35:1112–1118. 2017. View Article : Google Scholar

191 

Challita-Eid PM, Satpayev D, Yang P, An Z, Morrison K, Shostak Y, Raitano A, Nadell R, Liu W, Lortie DR, et al: Enfortumab vedotin antibody-drug conjugate targeting Nectin-4 is a highly potent therapeutic agent in multiple preclinical cancer models. Cancer Res. 76:3003–3013. 2016. View Article : Google Scholar : PubMed/NCBI

192 

Bardia A, Mayer IA, Vahdat LT, Tolaney SM, Isakoff SJ, Diamond JR, O'Shaughnessy J, Moroose RL, Santin AD, Abramson VG, et al: Sacituzumab govitecanhziy in refractory metastatic triple-negative breast cancer. N Engl J Med. 380:741–751. 2019. View Article : Google Scholar : PubMed/NCBI

193 

Raje N, Berdeja J, Lin Y, Siegel D, Jagannath S, Madduri D, Liedtke M, Rosenblatt J, Maus MV, Turka A, et al: Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med. 380:1726–1737. 2019. View Article : Google Scholar : PubMed/NCBI

194 

Merlino G, Fiascarelli A, Bigioni M, Bressan A, Carrisi C, Bellarosa D, Salerno M, Bugianesi R, Manno R, Bernadó Morales C, et al: MEN1309/OBT076, a first-in-class antibody-drug conjugate targeting CD205 in solid tumors. Mol Cancer Ther. 18:1533–1543. 2019. View Article : Google Scholar : PubMed/NCBI

195 

Zhan X, Wang B, Li Z, Li J, Wang H, Chen L, Jiang H, Wu M, Xiao J, Peng X, et al: Phase I trial of Claudin 18.2-specific chimeric antigen receptor T cells for advanced gastric and pancreatic adenocarcinoma. J Clin Oncol. 37(15 Suppl): S25092019. View Article : Google Scholar

196 

García-Alonso S, Ocaña A and Pandiella A: Resistance to antibody-drug conjugates. Cancer Res. 78:2159–2165. 2018. View Article : Google Scholar : PubMed/NCBI

197 

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

198 

Singal G, Miller PG, Agarwala V, Li G, Kaushik G, Backenroth D, Gossai A, Frampton GM, Torres AZ, Lehnert EM, et al: Association of patient characteristics and tumor genomics with clinical outcomes among patients with non-small cell lung cancer using a clinicogenomic database. JAMA. 321:1391–1399. 2019. View Article : Google Scholar : PubMed/NCBI

199 

Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, Barron DA, Zehir A, Jordan EJ, Omuro A, et al: Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 51:202–206. 2019. View Article : Google Scholar : PubMed/NCBI

200 

Rubinstein JC, Nicolson NG and Ahuja N: Next-generation sequencing in the management of gastric and esophageal cancers. Surg Clin North Am. 99:511–527. 2019. View Article : Google Scholar : PubMed/NCBI

201 

Hirsch FR, Suda K, Wiens J and Bunn PA Jr: New and emerging targeted treatments in advanced non-small-cell lung cancer. Lancet. 388:1012–1024. 2016. View Article : Google Scholar : PubMed/NCBI

202 

Rizvi H, Sanchez-Vega F, La K, Chatila W, Jonsson P, Halpenny D, Plodkowski A, Long N, Sauter JL, Rekhtman N, et al: Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung cancer profiled with targeted next-generation sequencing. J Clin Oncol. 36:633–641. 2018. View Article : Google Scholar : PubMed/NCBI

203 

Offin M, Rizvi H, Tenet M, Ni A, Sanchez-Vega F, Li BT, Drilon A, Kris MG, Rudin CM, Schultz N, et al: Tumor mutation burden and efficacy of EGFR-tyrosine kinase inhibitors in patients with EGFR-mutant lung cancers. Clin Cancer Res. 25:1063–1069. 2019. View Article : Google Scholar

204 

Buchhalter I, Rempel E, Endris V, Allgäuer M, Neumann O, Volckmar AL, Kirchner M, Leichsenring J, Lier A, von Winterfeld M, et al: Size matters: Dissecting key parameters for panel-based tumor mutational burden analysis. Int J Cancer. 144:848–858. 2019. View Article : Google Scholar

205 

Schneider G: Automating drug discovery. Nat Rev Drug Discov. 17:97–113. 2018. View Article : Google Scholar

206 

Katoh M: Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat Rev Clin Oncol. 16:105–122. 2019. View Article : Google Scholar

207 

Somashekhar SP, Sepúlveda MJ, Puglielli S, Norden AD, Shortliffe EH, Rohit Kumar C, Rauthan A, Arun Kumar N, Patil P, Rhee K, et al: Watson for oncology and breast cancer treatment recommendations: Agreement with an expert multidisciplinary tumor board. Ann Oncol. 29:418–423. 2018. View Article : Google Scholar : PubMed/NCBI

208 

Paley S and Karp PD: The MultiOmics explainer: Explaining omics results in the context of a pathway/genome database. BMC Bioinformatics. 20:3992019. View Article : Google Scholar : PubMed/NCBI

209 

Cheng JZ, Ni D, Chou YH, Qin J, Tiu CM, Chang YC, Huang CS, Shen D and Chen CM: Computer-aided diagnosis with deep learning architecture: Applications to breast lesions in US images and pulmonary nodules in CT scans. Sci Rep. 6:244542016. View Article : Google Scholar : PubMed/NCBI

210 

Esteva A, Kuprel B, Novoa RA, Ko J, Swetter SM, Blau HM and Thrun S: Dermatologist-level classification of skin cancer with deep neural networks. Nature. 542:115–118. 2017. View Article : Google Scholar : PubMed/NCBI

211 

Ardila D, Kiraly AP, Bharadwaj S, Choi B, Reicher JJ, Peng L, Tse D, Etemadi M, Ye W, Corrado G, et al: End-to-end lung cancer screening with three-dimensional deep learning on low-dose chest computed tomography. Nat Med. 25:954–961. 2019. View Article : Google Scholar : PubMed/NCBI

212 

Dascalu A and David EO: Skin cancer detection by deep learning and sound analysis algorithms: A prospective clinical study of an elementary dermoscope. EBioMedicine. 43:107–113. 2019. View Article : Google Scholar : PubMed/NCBI

213 

Mori Y, Kudo SE, Misawa M, Saito Y, Ikematsu H, Hotta K, Ohtsuka K, Urushibara F, Kataoka S, Ogawa Y, et al: Real-time use of artificial intelligence in identification of diminutive polyps during colonoscopy: A prospective study. Ann Intern Med. 169:357–366. 2018. View Article : Google Scholar : PubMed/NCBI

214 

Aboutalib SS, Mohamed AA, Berg WA, Zuley ML, Sumkin JH and Wu S: Deep learning to distinguish recalled but benign mammography images in breast cancer screening. Clin Cancer Res. 24:5902–5909. 2018. View Article : Google Scholar : PubMed/NCBI

215 

Coudray N, Ocampo PS, Sakellaropoulos T, Narula N, Snuderl M, Fenyö D, Moreira AL, Razavian N and Tsirigos A: Classification and mutation prediction from non-small cell lung cancer histopathology images using deep learning. Nat Med. 24:1559–1567. 2018. View Article : Google Scholar : PubMed/NCBI

216 

Kather JN, Pearson AT, Halama N, Jäger D, Krause J, Loosen SH, Marx A, Boor P, Tacke F, Neumann UP, et al: Deep learning can predict microsatellite instability directly from histology in gastrointestinal cancer. Nat Med. 25:1054–1056. 2019. View Article : Google Scholar : PubMed/NCBI

217 

Campanella G, Hanna MG, Geneslaw L, Miraflor A, Werneck Krauss, Silva V, Busam KJ, Brogi E, Reuter VE, Klimstra DS and Fuchs TJ: Clinical-grade computational pathology using weakly supervised deep learning on whole slide images. Nat Med. 25:1301–1309. 2019. View Article : Google Scholar : PubMed/NCBI

218 

Hekler A, Utikal JS, Enk AH, Solass W, Schmitt M, Klode J, Schadendorf D, Sondermann W, Franklin C, Bestvater F, et al: Deep learning outperformed 11 pathologists in the classification of histopathological melanoma images. Eur J Cancer. 118:91–96. 2019. View Article : Google Scholar : PubMed/NCBI

219 

Topol EJ: High-performance medicine: The convergence of human and artificial intelligence. Nat Med. 25:44–56. 2019. View Article : Google Scholar : PubMed/NCBI

220 

Zheng H, Bae Y, Kasimir-Bauer S, Tang R, Chen J, Ren G, Yuan M, Esposito M, Li W, Wei Y, et al: Therapeutic antibody targeting tumor- and osteoblastic niche-derived Jagged1 sensitizes bone metastasis to chemotherapy. Cancer Cell. 32:731–747.e6. 2017. View Article : Google Scholar : PubMed/NCBI

221 

Li DD, Zhao CH, Ding HW, Wu Q, Ren TS, Wang J, Chen CQ and Zhao QC: A novel inhibitor of ADAM17 sensitizes colorectal cancer cells to 5-Fluorouracil by reversing Notch and epithelial-mesenchymal transition in vitro and in vivo. Cell Prolif. 51:e124802018. View Article : Google Scholar : PubMed/NCBI

222 

Weber D, Lehal R, Frismantas V, Bourquin J, Bauer M, Murone M and Radtke F: 411P-Pharmacological activity of CB-103-an oral pan-NOTCH inhibitor with a novel mode of action. Ann Oncol. 28(Suppl 5): v122–v141. 2017. View Article : Google Scholar

223 

Moellering RE, Cornejo M, Davis TN, Del Bianco C, Aster JC, Blacklow SC, Kung AL, Gilliland DG, Verdine GL and Bradner JE: Direct inhibition of the NOTCH transcription factor complex. Nature. 462:182–188. 2009. View Article : Google Scholar : PubMed/NCBI

224 

Sano R, Krytska K, Larmour CE, Raman P, Martinez D, Ligon GF, Lillquist JS, Cucchi U, Orsini P, Rizzi S, et al: An antibody-drug conjugate directed to the ALK receptor demonstrates efficacy in preclinical models of neuroblastoma. Sci Transl Med. 11:pii: eaau9732. 2019. View Article : Google Scholar : PubMed/NCBI

225 

Boshuizen J, Koopman LA, Krijgsman O, Shahrabi A, van den Heuvel EG, Ligtenberg MA, Vredevoogd DW, Kemper K, Kuilman T, Song JY, et al: Cooperative targeting of melanoma heterogeneity with an AXL antibody-drug conjugate and BRAF/MEK inhibitors. Nat Med. 24:203–212. 2018. View Article : Google Scholar : PubMed/NCBI

226 

Tao Y, Wang R, Lai Q, Wu M, Wang Y, Jiang X, Zeng L, Zhou S, Li Z, Yang T, et al: Targeting of DDR1 with antibody-drug conjugates has antitumor effects in a mouse model of colon carcinoma. Mol Oncol. 13:1855–1873. 2019. View Article : Google Scholar : PubMed/NCBI

227 

Sommer A, Kopitz C, Schatz CA, Nising CF, Mahlert C, Lerchen HG, Stelte-Ludwig B, Hammer S, Greven S, Schuhmacher J, et al: Preclinical efficacy of the auristatin-based antibody-drug conjugate BAY 1187982 for the treatment of FGFR2-positive solid tumors. Cancer Res. 76:6331–6339. 2016. View Article : Google Scholar : PubMed/NCBI

228 

Surguladze D, Pennello A, Ren X, Mack T, Rigby A, Balderes P, Navarro E, Amaladas N, Eastman S, Topper M, et al: LY3076226, a novel anti-FGFR3 antibody drug conjugate exhibits potent and durable anti-tumor activity in tumor models harboring FGFR3 mutations or fusions. Cancer Res. 79(13 Suppl): S48352019.

229 

Rudra-Ganguly N, Challita-Eid PM, Lowe C, Mattie M, Moon SJ, Mendelsohn BA, Leavitt M, Virata C, A Verlinsky A, Capo L, et al: AGS62P1, a novel site-specific antibody drug conjugate targeting FLT3 exhibits potent anti-tumor activity regardless of FLT3 kinase activation status. Cancer Res. 76(14 Suppl): S5742016.

230 

Avilés P, Domínguez JM, Guillén MJ, Muñoz-Alonso MJ, Mateo C, Rodriguez-Acebes R, Molina-Guijarro JM, Francesch A, Martínez-Leal JF, Munt S, et al: MI130004, a novel antibody-drug conjugate combining trastuzumab with a molecule of marine origin, shows outstanding in iivo activity against HER2-expressing tumors. Mol Cancer Ther. 17:786–794. 2018. View Article : Google Scholar

231 

Koganemaru S, Kuboki Y, Koga Y, Kojima T, Yamauchi M, Maeda N, Kagari T, Hirotani K, Yasunaga M, Matsumura Y and Doi T: U3-1402, a novel HER3-targeting antibody-drug conjugate, for the treatment of colorectal cancer. Mol Cancer Ther. 18:2043–2050. 2019. View Article : Google Scholar : PubMed/NCBI

232 

Abrams T, Connor A, Fanton C, Cohen SB, Huber T, Miller K, Hong EE, Niu X, Kline J, Ison-Dugenny M, et al: Preclinical antitumor activity of a novel anti-c-KIT antibody-drug conjugate against mutant and wild-type c-KIT-positive solid tumors. Clin Cancer Res. 24:4297–4308. 2018. View Article : Google Scholar : PubMed/NCBI

233 

Strickler JH, Weekes CD, Nemunaitis J, Ramanathan RK, Heist RS, Morgensztern D, Angevin E, Bauer TM, Yue H, Motwani M, et al: First-in-human phase I, dose-escalation and -expansion study of telisotuzumab vedotin, an antibody-drug conjugate targeting c-Met, in patients with advanced solid tumors. J Clin Oncol. 36:3298–3306. 2018. View Article : Google Scholar : PubMed/NCBI

234 

Sachdev JC, Maitland ML, Sharma M, Moreno V, Boni V, Kummar S, Stringer-Reasor EM, Forero-Torres A, Lakhani NJ, Gibson B, et al: PF-06647020 (PF-7020), an antibody-drug conjugate (ADC) targeting protein tyrosine kinase 7 (PTK7), in patients (pts) with advanced solid tumors: Results of a phase I dose escalation and expansion study. J Clin Oncol. 36(15-Suppl): S55652018. View Article : Google Scholar

235 

Nguyen M, Miyakawa S, Kato J, Mori T, Arai T, Armanini M, Gelmon K, Yerushalmi R, Leung S, Gao D, et al: Preclinical efficacy and safety assessment of an antibody-drug conjugate targeting the c-RET proto-oncogene for breast carcinoma. Clin Cancer Res. 21:5552–5562. 2015. View Article : Google Scholar : PubMed/NCBI

236 

Yao HP, Feng L, Suthe SR, Chen LH, Weng TH, Hu CY, Jun ES, Wu ZG, Wang WL, Kim SC, et al: Therapeutic efficacy, pharmacokinetic profiles, and toxicological activities of humanized antibody-drug conjugate Zt/g4-MMAE targeting RON receptor tyrosine kinase for cancer therapy. J Immunother Cancer. 7:752019. View Article : Google Scholar : PubMed/NCBI

237 

Berger C, Sommermeyer D, Hudecek M, Berger M, Balakrishnan A, Paszkiewicz PJ, Kosasih PL, Rader C and Riddell SR: Safety of targeting ROR1 in primates with chimeric antigen receptor-modified T cells. Cancer Immunol Res. 3:206–216. 2015. View Article : Google Scholar :

238 

Trudel S, Lendvai N, Popat R, Voorhees PM, Reeves B, Libby EN, Richardson PG, Hoos A, Gupta I, Bragulat V, et al: Antibody-drug conjugate, GSK2857916, in relapsed/refractory multiple myeloma: An update on safety and efficacy from dose expansion phase I study. Blood Cancer J. 9:372019. View Article : Google Scholar : PubMed/NCBI

239 

Godwin CD, Gale RP and Walter RB: Gemtuzumab ozogamicin in acute myeloid leukemia. Leukemia. 31:1855–1868. 2017. View Article : Google Scholar : PubMed/NCBI

240 

Socinski MA, Kaye FJ, Spigel DR, Kudrik FJ, Ponce S, Ellis PM, Majem M, Lorigan P, Gandhi L, Gutierrez ME, et al: Phase 1/2 study of the CD56-targeting antibody-drug conjugate lorvotuzumab mertansine (IMGN901) in combination with carboplatin/etoposide in small-cell lung cancer patients with extensive-stage disease. Clin Lung Cancer. 18:68–76.e2. 2017. View Article : Google Scholar : PubMed/NCBI

241 

de Bono JS, Concin N, Hong DS, Thistlethwaite FC, Machiels JP, Arkenau HT, Plummer R, Jones RH, Nielsen D, Windfeld K, et al: Tisotumab vedotin in patients with advanced or metastatic solid tumours (InnovaTV 201): A first-in-human, multicentre, phase 1-2 trial. Lancet Oncol. 20:383–393. 2019. View Article : Google Scholar : PubMed/NCBI

242 

Dotan E, Cohen SJ, Starodub AN, Lieu CH, Messersmith WA, Simpson PS, Guarino MJ, Marshall JL, Goldberg RM, Hecht JR, et al: Phase I/II trial of labetuzumab govitecan (anti-CEACAM5/SN-38 antibody-drug conjugate) in patients with refractory or relapsing metastatic colorectal cancer. J Clin Oncol. 35:3338–3346. 2017. View Article : Google Scholar : PubMed/NCBI

243 

Zhu G, Foletti D, Liu X, Ding S, Melton Witt J, Hasa-Moreno A, Rickert M, Holz C, Aschenbrenner L, Yang AH, et al: Targeting CLDN18.2 by CD3 bispecific and ADC modalities for the treatments of gastric and pancreatic cancer. Sci Rep. 9:84202019. View Article : Google Scholar : PubMed/NCBI

244 

Bhakta S, Crocker LM, Chen Y, Hazen M, Schutten MM, Li D, Kuijl C, Ohri R, Zhong F, Poon KA, et al: An anti-GDNF family receptor alpha 1 (GFRA1) antibody-drug conjugate for the treatment of hormone receptor-positive breast cancer. Mol Cancer Ther. 17:638–649. 2018. View Article : Google Scholar

245 

Ott PA, Pavlick AC, Johnson DB, Hart LL, Infante JR, Luke JJ, Lutzky J, Rothschild NE, Spitler LE, Cowey CL, et al: A phase 2 study of glembatumumab vedotin, an antibody-drug conjugate targeting glycoprotein NMB, in patients with advanced melanoma. Cancer. 125:1113–1123. 2019. View Article : Google Scholar : PubMed/NCBI

246 

Gong X, Azhdarinia A, Ghosh SC, Xiong W, An Z, Liu Q and Carmon KS: LGR5-targeted antibody-drug conjugate eradicates gastrointestinal tumors and prevents recurrence. Mol Cancer Ther. 15:1580–1590. 2016. View Article : Google Scholar : PubMed/NCBI

247 

Purcell JW, Tanlimco SG, Hickson J, Fox M, Sho M, Durkin L, Uziel T, Powers R, Foster K, McGonigal T, et al: LRRC15 is a novel mesenchymal protein and stromal target for antibody-drug conjugates. Cancer Res. 78:4059–4072. 2018. View Article : Google Scholar : PubMed/NCBI

248 

Willuda J, Linden L, Lerchen HG, Kopitz C, Stelte-Ludwig B, Pena C, Lange C, Golfier S, Kneip C, Carrigan PE, et al: Preclinical antitumor efficacy of BAY 1129980-a novel auristatin-based anti-C4.4A (LYPD3) antibody-drug conjugate for the treatment of non-small cell lung cancer. Mol Cancer Ther. 16:893–904. 2017. View Article : Google Scholar : PubMed/NCBI

249 

Golfier S, Kopitz C, Kahnert A, Heisler I, Schatz CA, Stelte-Ludwig B, Mayer-Bartschmid A, Unterschemmann K, Bruder S, Linden L, et al: Anetumab ravtansine: A novel mesothelin-targeting antibody-drug conjugate cures tumors with heterogeneous target expression favored by bystander effect. Mol Cancer Ther. 13:1537–1548. 2014. View Article : Google Scholar : PubMed/NCBI

250 

Banerjee S, Oza AM, Birrer MJ, Hamilton EP, Hasan J, Leary A, Moore KN, Mackowiak-Matejczyk B, Pikiel J, Ray-Coquard I, et al: Anti-NaPi2b antibody-drug conjugate lifastuzumab vedotin (DNIB0600A) compared with pegylated liposomal doxorubicin in patients with platinum-resistant ovarian cancer in a randomized, open-label, phase II study. Ann Oncol. 29:917–923. 2018. View Article : Google Scholar : PubMed/NCBI

251 

Sussman D, Smith LM, Anderson ME, Duniho S, Hunter JH, Kostner H, Miyamoto JB, Nesterova A, Westendorf L, Van Epps HA, et al: SGN-LIV1A: A novel antibody-drug conjugate targeting LIV-1 for the treatment of metastatic breast cancer. Mol Cancer Ther. 13:2991–3000. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2020
Volume 45 Issue 2

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: Precision medicine for human cancers with Notch signaling dysregulation (Review). Int J Mol Med 45: 279-297, 2020
APA
Katoh, M., & Katoh, M. (2020). Precision medicine for human cancers with Notch signaling dysregulation (Review). International Journal of Molecular Medicine, 45, 279-297. https://doi.org/10.3892/ijmm.2019.4418
MLA
Katoh, M., Katoh, M."Precision medicine for human cancers with Notch signaling dysregulation (Review)". International Journal of Molecular Medicine 45.2 (2020): 279-297.
Chicago
Katoh, M., Katoh, M."Precision medicine for human cancers with Notch signaling dysregulation (Review)". International Journal of Molecular Medicine 45, no. 2 (2020): 279-297. https://doi.org/10.3892/ijmm.2019.4418